1
|
Wu B, Cheng Z, Li X, Liang M, Wang X, Pi D, Liu J, Li H, Zhao J, Wang J, Liang F, Wang X. The developmental toxicity of bisphenol F exposure on the zebrafish larvae. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 298:118282. [PMID: 40344781 DOI: 10.1016/j.ecoenv.2025.118282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 05/03/2025] [Accepted: 05/04/2025] [Indexed: 05/11/2025]
Abstract
As a major substitute for the bisphenol A (BPA), the use of the bisphenol F (BPF) has increased dramatically in recent years. Growing evidence suggest that BPF shares numerous toxicological properties with BPA, raising the concerns about its potential impact on the health of organisms. However, the developmental toxicity of BPF remains poorly understood. In this study, we conducted a 5-day BPF exposure experiment on zebrafish (Danio rerio) from blastula stage at concentrations of 2, 20, 200, and 2000 µg/L. Our results demonstrated a significant increase in hatching rates across all treatment groups at 2 days post-fertilization (dpf). The esr1 was significantly upregulated at 2000 µg/L by 5 dpf, while no significant change was observed in ar. The frequency of operculum loss significantly increased at exposure concentrations of 20, 200, and 2000 µg/L, and a notable increase in notochord loss was observed at 2000 µg/L. To explore the underlying mechanisms, transcriptomic analysis was performed to identify differentially expressed genes (DEGs). GO and KEGG pathway enrichment analysis revealed that the toxic effects of BPF were closely associated with osteoclast differentiation, the FoxO signaling pathway, and the MAPK signaling pathway. These pathways influenced critical biological processes, including response to stimuli, animal organ morphogenesis, detoxification, and biomineralization. This study provides evidence that BPF exposure at environmentally relevant concentrations (2 µg/L) is harmful to hatching, concentrations above 20 µg/L exhibit estrogenic-disrupting activity and exert toxicological effects on the development of the head skeleton in zebrafish. These effects are particularly linked to disruptions in osteoclast differentiation.
Collapse
Affiliation(s)
- Biyu Wu
- Institute of Modern Aquaculture Science and Engineering, Guangdong-Macao Joint Laboratory for Aquaculture Breeding Development and Innovation, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Zirui Cheng
- Institute of Modern Aquaculture Science and Engineering, Guangdong-Macao Joint Laboratory for Aquaculture Breeding Development and Innovation, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Xiang Li
- Institute of Modern Aquaculture Science and Engineering, Guangdong-Macao Joint Laboratory for Aquaculture Breeding Development and Innovation, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Minxing Liang
- Institute of Modern Aquaculture Science and Engineering, Guangdong-Macao Joint Laboratory for Aquaculture Breeding Development and Innovation, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Xue Wang
- Institute of Modern Aquaculture Science and Engineering, Guangdong-Macao Joint Laboratory for Aquaculture Breeding Development and Innovation, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Duan Pi
- Institute of Modern Aquaculture Science and Engineering, Guangdong-Macao Joint Laboratory for Aquaculture Breeding Development and Innovation, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Jiayi Liu
- Institute of Modern Aquaculture Science and Engineering, Guangdong-Macao Joint Laboratory for Aquaculture Breeding Development and Innovation, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Huiling Li
- Institute of Modern Aquaculture Science and Engineering, Guangdong-Macao Joint Laboratory for Aquaculture Breeding Development and Innovation, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Jun Zhao
- Institute of Modern Aquaculture Science and Engineering, Guangdong-Macao Joint Laboratory for Aquaculture Breeding Development and Innovation, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Junjie Wang
- Institute of Modern Aquaculture Science and Engineering, Guangdong-Macao Joint Laboratory for Aquaculture Breeding Development and Innovation, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Fang Liang
- Institute of Modern Aquaculture Science and Engineering, Guangdong-Macao Joint Laboratory for Aquaculture Breeding Development and Innovation, School of Life Sciences, South China Normal University, Guangzhou 510631, China.
| | - Xuegeng Wang
- Institute of Modern Aquaculture Science and Engineering, Guangdong-Macao Joint Laboratory for Aquaculture Breeding Development and Innovation, School of Life Sciences, South China Normal University, Guangzhou 510631, China.
| |
Collapse
|
2
|
André DCA, Oliveira PF, Alves MG, Martins AD. Caloric Restriction and Sirtuins as New Players to Reshape Male Fertility. Metabolites 2025; 15:303. [PMID: 40422880 DOI: 10.3390/metabo15050303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 04/25/2025] [Accepted: 04/30/2025] [Indexed: 05/28/2025] Open
Abstract
Over the years, caloric intake has remained a subject of profound scrutiny. Within the scientific community, there has been rigorous debate to ascertain which path is most ideal for enhancing quality of life and extending the human lifespan. Caloric restriction has been shown to be a promising contributor towards longevity and delaying the onset of age-related diseases. This diet consists of a reduction in caloric intake while maintaining essential energy and nutritional requirements to achieve optimal health while avoiding malnutrition. However, the effects of this nutritional regimen on male reproductive health have not yet been comprehensively studied. Nevertheless, such a complex process will certainly be regulated by a variety of metabolic sensors, likely sirtuins. Evidence has been gathered regarding this group of enzymes, and their ability to regulate processes such as chromatin condensation, the cell cycle, insulin signaling, and glucose and lipid metabolism, among many others. Concerning testicular function and male fertility, sirtuins can modulate certain metabolic processes through their interaction with the hypothalamic-pituitary-gonadal axis and mitochondrial dynamics, among many others, which remain largely unexplored. This review explores the impact of caloric restriction on male fertility, highlighting the emerging role of sirtuins as key regulators of male reproductive health through their influence on cellular metabolism.
Collapse
Affiliation(s)
- Diana C A André
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Pedro F Oliveira
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Marco G Alves
- Institute of Biomedicine, Department of Medical Sciences (iBiMED), University of Aveiro, 3810-193 Aveiro, Portugal
| | - Ana D Martins
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| |
Collapse
|
3
|
Stevens CM, Weeks K, Jain SK. Potential of Vitamin D and l-Cysteine Co-supplementation to Downregulate Mammalian Target of Rapamycin: A Novel Therapeutic Approach to Diabetes. Metab Syndr Relat Disord 2025; 23:13-22. [PMID: 39279596 PMCID: PMC12021770 DOI: 10.1089/met.2024.0146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/18/2024] Open
Abstract
Diabetes, a metabolic disease associated with an increased health care burden and mortality, is currently on the rise. Both upregulation of the mammalian target of rapamycin (mTOR) and decreased levels of vitamin D (VD) and l-cysteine (LC) have been associated with diabetes. The overactivation of mTOR leads to insulin desensitization and metabolic dysfunction including uncontrolled hyperglycemia. This review summarizes various studies that have shown an inhibitory effect of VD or LC on mTOR activity. Findings from preclinical studies suggest that optimizing the VD and LC status in patients with diabetes can result in mTOR suppression, which has the potential to protect these individuals from microvascular and macrovascular complications while enhancing the regulation of their blood glucose. Given this information, finding ways to suppress mTOR signaling and also increasing VD and LC status is a possible therapeutic approach that might aid patients with diabetes. Future clinical trials are needed to investigate whether VD and LC co-supplementation can successfully downregulate mTOR and can be used as adjuvant therapy in patients with diabetes.
Collapse
Affiliation(s)
- Christopher M. Stevens
- Departments of Pediatrics and Medicine, Louisiana State University Health Sciences Center, Shreveport, Louisiana, USA
| | - Kathrine Weeks
- Department of Chemistry, Centenary College of Louisiana, Shreveport, Louisiana, USA
| | - Sushil K. Jain
- Departments of Pediatrics and Medicine, Louisiana State University Health Sciences Center, Shreveport, Louisiana, USA
| |
Collapse
|
4
|
Reis IA, Baldassini WA, Ramírez-Zamudio GD, de Farias IMSC, Chiaratti MR, Pereira Junior S, Nociti RP, Carvalho PHV, Curi RA, Pereira GL, Chardulo LAL, Neto ORM. Muscle tissue transcriptome of F1 Angus-Nellore bulls and steers feedlot finished: impacts on intramuscular fat deposition. BMC Genomics 2024; 25:1178. [PMID: 39633259 PMCID: PMC11616301 DOI: 10.1186/s12864-024-11066-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 11/18/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND Castration is a common practice in beef cattle production systems to manage breeding and enhance meat quality by promoting intramuscular fat (IMF) deposition, known as marbling. However, the molecular mechanisms that are influenced by castration in beef cattle are poorly understood. The aim of this study was to identify differentially expressed genes (DEGs) and metabolic pathways that regulate IMF deposition in crossbred cattle by RNA sequencing (RNA-Seq) of skeletal muscle tissue. Six hundred and forty F1 Angus-Nellore bulls and steers (n = 320/group) were submitted to feedlot finishing for 180 days. Sixty Longissimus thoracis muscle samples were collected randomly from each group in the hot carcass (at slaughter) and 48 h post-mortem (at deboning), at between 12th and 13th thoracic vertebrae. Three muscle samples of each group were randomly selected for RNA-Seq analysis, while the post-deboning meat samples were submitted to determination of IMF content. RESULTS Steers had a 2.7-fold greater IMF content than bulls (5.59 vs. 2.07%; P < 0.01). A total of 921 DEGs (FDR < 0.05) were identified in contrast between Bulls versus Steers; of these, 371 were up-regulated, and 550 were down-regulated. Functional transcriptome enrichment analysis revealed differences in biological processes and metabolic pathways related to adipogenesis and lipogenesis, such as insulin resistance, AMPK, cAMP, regulation of lipolysis in adipocytes, and PI3K-Akt signaling pathways. Candidate genes such as FOXO1, PPARG, PCK2, CALM1, LEP, ADIPOQ, FASN, FABP4, PLIN1, PIK3R3, ROCK2, ADCY5, and ADORA1 were regulated in steers, which explains the expressive difference in IMF content when compared to bulls. CONCLUSIONS The current findings suggest the importance of these pathways and genes for lipid metabolism in steers with greater IMF. Notably, this study reveals for the first time the involvement of the PI3K-Akt pathway and associated genes in regulating IMF deposition in F1 Angus-Nellore cattle. Castration influenced DEGs linked to energy metabolism and lipid biosynthesis, highlighting key molecular players responsible for IMF accumulation post-castration in beef cattle.
Collapse
Affiliation(s)
- Irene Alexandre Reis
- College of Agriculture and Veterinary Sciences (FCAV), Department of Animal Science, São Paulo State University "Júlio de Mesquita Filho" (UNESP), Jaboticabal, SP, 14884-900, Brazil
| | - Welder Angelo Baldassini
- College of Agriculture and Veterinary Sciences (FCAV), Department of Animal Science, São Paulo State University "Júlio de Mesquita Filho" (UNESP), Jaboticabal, SP, 14884-900, Brazil
- College of Veterinary Medicine and Animal Science (FMVZ), São Paulo State University (UNESP), Botucatu, SP, 18618-681, Brazil
| | | | - Iasmin Myrele Santos Calaça de Farias
- College of Agriculture and Veterinary Sciences (FCAV), Department of Animal Science, São Paulo State University "Júlio de Mesquita Filho" (UNESP), Jaboticabal, SP, 14884-900, Brazil
| | - Marcos Roberto Chiaratti
- Department of Genetics and Evolution, Federal University of São Carlos (UFSCar), São Carlos, SP, 13565-905, Brazil
| | - Sérgio Pereira Junior
- Department of Genetics and Evolution, Federal University of São Carlos (UFSCar), São Carlos, SP, 13565-905, Brazil
| | - Ricardo Perecin Nociti
- College of Animal Science and Foods Engineering, University of São Paulo, Pirassununga, SP, 13635-900, Brazil
| | | | - Rogério Abdallah Curi
- College of Agriculture and Veterinary Sciences (FCAV), Department of Animal Science, São Paulo State University "Júlio de Mesquita Filho" (UNESP), Jaboticabal, SP, 14884-900, Brazil
- College of Veterinary Medicine and Animal Science (FMVZ), São Paulo State University (UNESP), Botucatu, SP, 18618-681, Brazil
| | - Guilherme Luis Pereira
- College of Agriculture and Veterinary Sciences (FCAV), Department of Animal Science, São Paulo State University "Júlio de Mesquita Filho" (UNESP), Jaboticabal, SP, 14884-900, Brazil
- College of Veterinary Medicine and Animal Science (FMVZ), São Paulo State University (UNESP), Botucatu, SP, 18618-681, Brazil
| | - Luis Artur Loyola Chardulo
- College of Agriculture and Veterinary Sciences (FCAV), Department of Animal Science, São Paulo State University "Júlio de Mesquita Filho" (UNESP), Jaboticabal, SP, 14884-900, Brazil
- College of Veterinary Medicine and Animal Science (FMVZ), São Paulo State University (UNESP), Botucatu, SP, 18618-681, Brazil
| | - Otávio Rodrigues Machado Neto
- College of Agriculture and Veterinary Sciences (FCAV), Department of Animal Science, São Paulo State University "Júlio de Mesquita Filho" (UNESP), Jaboticabal, SP, 14884-900, Brazil.
- College of Veterinary Medicine and Animal Science (FMVZ), São Paulo State University (UNESP), Botucatu, SP, 18618-681, Brazil.
| |
Collapse
|
5
|
Yu W, Tong MJ, Wu GH, Ma TL, Cai CD, Wang LP, Zhang YK, Gu JL, Yan ZQ. FoxO3 Regulates Mouse Bone Mesenchymal Stem Cell Fate and Bone-Fat Balance During Skeletal Aging. Stem Cells Dev 2024; 33:365-375. [PMID: 38661524 DOI: 10.1089/scd.2024.0055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2024] Open
Abstract
Age-related osteoporosis is characterized by an imbalance between osteogenic and adipogenic differentiation in bone mesenchymal stem cells (BMSCs). Forkhead box O 3 (FoxO3) transcription factor is involved in lifespan and cell differentiation. In this study, we explore whether FoxO3 regulates age-related bone loss and marrow fat accumulation. The expression levels of FoxO3 in BMSCs during aging were detected in vivo and in vitro. To explore the role of FoxO3 in osteogenic and adipogenic differentiation, primary BMSCs were isolated from young and aged mice. FoxO3 expression was modulated by adenoviral vector transfection. The role of FoxO3 in bone-fat balance was evaluated by alizarin red S staining, oil red O staining, quantitative reverse transcription-polymerase chain reaction, Western blot, and histological analysis. Age-related bone loss and fat deposit are associated with downregulation of FoxO3. Overexpression of FoxO3 alleviated age-related bone loss and marrow fat accumulation in aged mice. Mechanistically, FoxO3 reduced adipogenesis and enhanced osteogenesis of BMSCs via downregulation of PPAR-γ and Notch signaling, respectively. In conclusion, FoxO3 is an essential factor controlling the fate of BMSCs and is a potential target for the prevention of age-related osteoporosis.
Collapse
Affiliation(s)
- Wei Yu
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Min-Ji Tong
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Guo-Hao Wu
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tian-Le Ma
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chuan-Dong Cai
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Li-Peng Wang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ying-Kai Zhang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jin-Lun Gu
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zuo-Qin Yan
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
6
|
Lee HW, Karki R, Han JH. Inhibition of the RPS6KA1/FoxO1 signaling axis by hydroxycitric acid attenuates HFD-induced obesity through MCE suppression. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155551. [PMID: 38569293 DOI: 10.1016/j.phymed.2024.155551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 10/02/2023] [Accepted: 03/19/2024] [Indexed: 04/05/2024]
Abstract
BACKGROUND Because obesity is associated with a hyperplasia-mediated increase in adipose tissue, inhibiting cell proliferation during mitotic clonal expansion (MCE) is a leading strategy for preventing obesity. Although (-)-hydroxycitric acid (HCA) is used to control obesity, the molecular mechanisms underlying its effects on MCE are poorly understood. PURPOSE This study aimed to investigate the potential effects of HCA on MCE and underlying molecular mechanisms affecting adipogenesis and obesity improvements. METHODS Preadipocyte cell line, 3T3-L1, were treated with HCA; oil red O, cell proliferation, cell cycle, and related alterations in signaling pathways were examined. High-fat diet (HFD)-fed mice were administered HCA for 12 weeks; body and adipose tissues weights were evaluated, and the regulation of signaling pathways in epidydimal white adipose tissue were examined in vivo. RESULTS Here, we report that during MCE, HCA attenuates the proliferation of the preadipocyte cell line, 3T3-L1, by arresting the cell cycle at the G0/G1 phase. In addition, HCA markedly inhibits Forkhead Box O1 (FoxO1) phosphorylation, thereby inducing the expression of cyclin-dependent kinase inhibitor 1B and suppressing the levels of cyclin-dependent kinase 2, cyclin E1, proliferating cell nuclear antigen, and phosphorylated retinoblastoma. Importantly, we found that ribosomal protein S6 kinase A1 (RPS6KA1) influences HCA-mediated inactivation of FoxO1 and its nuclear exclusion. An animal model of obesity revealed that HCA reduced high-fat diet-induced obesity by suppressing adipocyte numbers as well as epididymal and mesenteric white adipose tissue mass, which is attributed to the regulation of RPS6KA1, FoxO1, CDKN1B and PCNA that had been consistently identified in vitro. CONCLUSIONS These findings provide novel insights into the mechanism by which HCA regulates adipogenesis and highlight the RPS6KA1/FoxO1 signaling axis as a therapeutic target for obesity.
Collapse
Affiliation(s)
- Hyung-Won Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Woosuk University, Wanju 55338, Republic of Korea
| | - Rajendra Karki
- Department of Biological Sciences, College of Natural Science, Seoul National University, Seoul 08826, South Korea; Nexus Institute of Research and Innovation (NIRI), Kathmandu, Nepal
| | - Joo-Hui Han
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Woosuk University, Wanju 55338, Republic of Korea.
| |
Collapse
|
7
|
Janssen AWF, Jansen Holleboom W, Rijkers D, Louisse J, Hoekstra SA, Schild S, Vrolijk MF, Hoogenboom RLAP, Beekmann K. Determination of in vitro immunotoxic potencies of a series of perfluoralkylsubstances (PFASs) in human Namalwa B lymphocyte and human Jurkat T lymphocyte cells. FRONTIERS IN TOXICOLOGY 2024; 6:1347965. [PMID: 38549690 PMCID: PMC10976438 DOI: 10.3389/ftox.2024.1347965] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 03/01/2024] [Indexed: 01/05/2025] Open
Abstract
Exposure to PFASs is associated to several adverse health effects, such as immunotoxicity. Immunotoxic effects of PFOA and PFOS, including a reduced antibody response in both experimental animals and humans, have been reported. However, there is limited understanding of the underlying mechanisms involved. Moreover, there is only a restricted amount of immunotoxicity data available for a limited number of PFASs. In the current study the effects of 15 PFASs, including short- and long-chain perfluorinated carboxylic and sulfonic acids, fluorotelomer alcohols, and perfluoralkyl ether carboxylic acids were studied on the expression of recombinant activating gene 1 (RAG1) and RAG2 in the Namalwa human B lymphoma cell line, and on the human IL-2 promotor activity in Jurkat T-cells. Concentration-response data were subsequently used to derive in vitro relative potencies through benchmark dose analysis. In vitro relative potency factors (RPFs) were obtained for 6 and 9 PFASs based on their effect on RAG1 and RAG2 gene expression in Namalwa B-cells, respectively, and for 10 PFASs based on their inhibitory effect on IL-2 promotor activity in Jurkat T-cells. The most potent substances were HFPO-TA for the reduction of RAG1 and RAG2 gene expression in Namalwa cells (RPFs of 2.1 and 2.3 respectively), and PFDA on IL-2 promoter activity (RPF of 9.1). RAG1 and RAG2 play a crucial role in V (D)J gene recombination, a process for acquiring a varied array of antibodies crucial for antigen recognition. Hence, the effects observed in Namalwa cells might indicate a PFAS-induced impairment of generating a diverse range of B-cells essential for antigen recognition. The observed outcomes in the Jurkat T-cells suggest a possible PFAS-induced reduction of T-cell activation, which may contribute to a decline in the T-cell dependent antibody response. Altogether, the present study provides potential mechanistic insights into the reported PFAS-induced decreased antibody response. Additionally, the presented in vitro models may represent useful tools for assessing the immunotoxic potential of PFASs and prioritization for further risk assessment.
Collapse
Affiliation(s)
- Aafke W. F. Janssen
- Wageningen Food Safety Research (WFSR), Wageningen University and Research, Wageningen, Netherlands
| | - Wendy Jansen Holleboom
- Wageningen Food Safety Research (WFSR), Wageningen University and Research, Wageningen, Netherlands
| | - Deborah Rijkers
- Wageningen Food Safety Research (WFSR), Wageningen University and Research, Wageningen, Netherlands
| | - Jochem Louisse
- Wageningen Food Safety Research (WFSR), Wageningen University and Research, Wageningen, Netherlands
- European Food Safety Authority, Parma, Italy
| | - Sjoerdtje A. Hoekstra
- Wageningen Food Safety Research (WFSR), Wageningen University and Research, Wageningen, Netherlands
| | - Sanne Schild
- Wageningen Food Safety Research (WFSR), Wageningen University and Research, Wageningen, Netherlands
| | - Misha F. Vrolijk
- Department of Pharmacology and Toxicology, Maastricht University, Maastricht, Netherlands
| | - Ron L. A. P. Hoogenboom
- Wageningen Food Safety Research (WFSR), Wageningen University and Research, Wageningen, Netherlands
| | - Karsten Beekmann
- Wageningen Food Safety Research (WFSR), Wageningen University and Research, Wageningen, Netherlands
| |
Collapse
|
8
|
Kim DH, Lee S, Noh SG, Lee J, Chung HY. FoxO6-mediated ApoC3 upregulation promotes hepatic steatosis and hyperlipidemia in aged rats fed a high-fat diet. Aging (Albany NY) 2024; 16:4095-4115. [PMID: 38441531 PMCID: PMC10968681 DOI: 10.18632/aging.205610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 01/24/2024] [Indexed: 03/22/2024]
Abstract
FoxO6, an identified factor, induces hyperlipidemia and hepatic steatosis during aging by activating hepatic lipoprotein secretion and lipogenesis leading to increased ApoC3 concentrations in the bloodstream. However, the intricate mechanisms underlying hepatic steatosis induced by elevated FoxO6 under hyperglycemic conditions remain intricate and require further elucidation. In order to delineate the regulatory pathway involving ApoC3 controlled by FoxO6 and its resultant functional impacts, we employed a spectrum of models including liver cell cultures, aged rats subjected to HFD, transgenic mice overexpressing FoxO6 (FoxO6-Tg), and FoxO6 knockout mice (FoxO6-KO). Our findings indicate that FoxO6 triggered ApoC3-driven lipid accumulation in the livers of aged rats on an HFD and in FoxO6-Tg, consequently leading to hepatic steatosis and hyperglycemia. Conversely, the absence of FoxO6 attenuated the expression of genes involved in lipogenesis, resulting in diminished hepatic lipid accumulation and mitigated hyperlipidemia in murine models. Additionally, the upregulation of FoxO6 due to elevated glucose levels led to increased ApoC3 expression, consequently instigating cellular triglyceride mediated lipid accumulation. The transcriptional activation of FoxO6 induced by both the HFD and high glucose levels resulted in hepatic steatosis by upregulating ApoC3 and genes associated with gluconeogenesis in aged rats and liver cell cultures. Our conclusions indicate that the upregulation of ApoC3 by FoxO6 promotes the development of hyperlipidemia, hyperglycemia, and hepatic steatosis in vivo, and in vitro. Taken together, our findings underscore the significance of FoxO6 in driving hyperlipidemia and hepatic steatosis specifically under hyperglycemic states by enhancing the expression of ApoC3 in aged rats.
Collapse
Affiliation(s)
- Dae Hyun Kim
- Department of Food Science and Technology, College of Natural Resources and Life Science, Pusan National University, Miryang-si, Gyeongsangnam-do 50463, Republic of Korea
| | - Seulah Lee
- Department of Pharmacy, Research Institute for Drug Development, College of Pharmacy, Pusan National University, Geumjeong-gu, Busan 46241, Republic of Korea
| | - Sang Gyun Noh
- Department of Pharmacy, Research Institute for Drug Development, College of Pharmacy, Pusan National University, Geumjeong-gu, Busan 46241, Republic of Korea
| | - Jaewon Lee
- Department of Pharmacy, Research Institute for Drug Development, College of Pharmacy, Pusan National University, Geumjeong-gu, Busan 46241, Republic of Korea
| | - Hae Young Chung
- Department of Pharmacy, Research Institute for Drug Development, College of Pharmacy, Pusan National University, Geumjeong-gu, Busan 46241, Republic of Korea
| |
Collapse
|
9
|
Guo X, Peng K, He Y, Xue L. Mechanistic regulation of FOXO transcription factors in the nucleus. Biochim Biophys Acta Rev Cancer 2024; 1879:189083. [PMID: 38309444 DOI: 10.1016/j.bbcan.2024.189083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 01/28/2024] [Accepted: 01/31/2024] [Indexed: 02/05/2024]
Abstract
FOXO proteins represent evolutionarily conserved transcription factors (TFs) that play critical roles in responding to various physiological signals or pathological stimuli, either through transcription-dependent or -independent mechanisms. Dysfunction of these proteins have been implicated in numerous diseases, including cancer. Although the regulation of FOXO TFs shuttling between the cytoplasm and the nucleus has been extensively studied and reviewed, there's still a lack of a comprehensive review focusing on the intricate interactions between FOXO, DNA, and cofactors in the regulation of gene expression. In this review, we aim to summarize recent advances and provide a detailed understanding of the mechanism underlying FOXO proteins binding to target DNA. Additionally, we will discuss the challenges associated with pharmacological approaches in modulating FOXO function, and explore the dynamic association between TF, DNA, and RNA on chromatin. This review will contribute to a better understanding of mechanistic regulations of eukaryotic TFs within the nucleus.
Collapse
Affiliation(s)
- Xiaowei Guo
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, Changsha, China; The Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Changsha, China.
| | - Kai Peng
- Institute of Intervention Vessel, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China
| | - Yanwen He
- Changsha Stomatological Hospital, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Lei Xue
- Institute of Intervention Vessel, Shanghai 10th People's Hospital, Shanghai Key Laboratory of Signaling and Diseases Research, School of Life Science and Technology, Tongji University, Shanghai, China.
| |
Collapse
|
10
|
Zhang Y, Wang Q, Xue H, Guo Y, Wei S, Li F, Gong L, Pan W, Jiang P. Epigenetic Regulation of Autophagy in Bone Metabolism. FUNCTION 2024; 5:zqae004. [PMID: 38486976 PMCID: PMC10935486 DOI: 10.1093/function/zqae004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 01/18/2024] [Accepted: 01/24/2024] [Indexed: 03/17/2024] Open
Abstract
The skeletal system is crucial for supporting bodily functions, protecting vital organs, facilitating hematopoiesis, and storing essential minerals. Skeletal homeostasis, which includes aspects such as bone density, structural integrity, and regenerative processes, is essential for normal skeletal function. Autophagy, an intricate intracellular mechanism for degrading and recycling cellular components, plays a multifaceted role in bone metabolism. It involves sequestering cellular waste, damaged proteins, and organelles within autophagosomes, which are then degraded and recycled. Autophagy's impact on bone health varies depending on factors such as regulation, cell type, environmental cues, and physiological context. Despite being traditionally considered a cytoplasmic process, autophagy is subject to transcriptional and epigenetic regulation within the nucleus. However, the precise influence of epigenetic regulation, including DNA methylation, histone modifications, and non-coding RNA expression, on cellular fate remains incompletely understood. The interplay between autophagy and epigenetic modifications adds complexity to bone cell regulation. This article provides an in-depth exploration of the intricate interplay between these two regulatory paradigms, with a focus on the epigenetic control of autophagy in bone metabolism. Such an understanding enhances our knowledge of bone metabolism-related disorders and offers insights for the development of targeted therapeutic strategies.
Collapse
Affiliation(s)
- Yazhou Zhang
- Department of Foot and Ankle Surgery, Tengzhou Central People’s Hospital, Tengzhou 277500, China
| | - Qianqian Wang
- Department of Pediatric Intensive Care Unit, Tengzhou Central People’s Hospital, Tengzhou 277500, China
| | - Hongjia Xue
- Department of Computer Science, University College London, London, WC1E 6BT, UK
| | - Yujin Guo
- Institute of Clinical Pharmacy & Pharmacology, Jining First People’s Hospital, Jining 272000, China
| | - Shanshan Wei
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250000, China
- Department of Graduate, Shandong Academy of Medical Sciences, Shandong First Medical University, Jinan 250000, China
| | - Fengfeng Li
- Department of Neurosurgery, Tengzhou Central People’s Hospital, Tengzhou 277500, China
| | - Linqiang Gong
- Department of Gastroenterology, Tengzhou Central People's Hospital, Tengzhou 277500, China
| | - Weiliang Pan
- Department of Foot and Ankle Surgery, Tengzhou Central People’s Hospital, Tengzhou 277500, China
| | - Pei Jiang
- Translational Pharmaceutical Laboratory, Jining First People’s Hospital, Shandong First Medical University, Jining 272000, China
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining 272000, China
| |
Collapse
|
11
|
Tokumasu R, Yasuhara R, Kang S, Funatsu T, Mishima K. Transcription factor FoxO1 regulates myoepithelial cell diversity and growth. Sci Rep 2024; 14:1069. [PMID: 38212454 PMCID: PMC10784559 DOI: 10.1038/s41598-024-51619-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 01/08/2024] [Indexed: 01/13/2024] Open
Abstract
Salivary gland myoepithelial cells regulate saliva secretion and have been implicated in the histological diversity of salivary gland tumors. However, detailed functional analysis of myoepithelial cells has not been determined owing to the few of the specific marker to isolate them. We isolated myoepithelial cells from the submandibular glands of adult mice using the epithelial marker EpCAM and the cell adhesion molecule CD49f as indicators and found predominant expression of the transcription factor FoxO1 in these cells. RNA-sequence analysis revealed that the expression of cell cycle regulators was negatively regulated in FoxO1-overexpressing cells. Chromatin immunoprecipitation analysis showed that FoxO1 bound to the p21/p27 promoter DNA, indicating that FoxO1 suppresses cell proliferation through these factors. In addition, FoxO1 induced the expression of ectodysplasin A (Eda) and its receptor Eda2r, which are known to be associated with X-linked hypohidrotic ectodermal dysplasia and are involved in salivary gland development in myoepithelial cells. FoxO1 inhibitors suppressed Eda/Eda2r expression and salivary gland development in primordial organ cultures after mesenchymal removal. Although mesenchymal cells are considered a source of Eda, myoepithelial cells might be one of the resources of Eda. These results suggest that FoxO1 regulates myoepithelial cell proliferation and Eda secretion during salivary gland development in myoepithelial cells.
Collapse
Affiliation(s)
- Rino Tokumasu
- Division of Pathology, Department of Oral Diagnostic Sciences, School of Dentistry, Showa University, Tokyo, 142-8555, Japan
- Division of Dentistry for Persons with Disabilities, Department of Perioperative Medicine, Graduate School of Dentistry, Showa University, Tokyo, 142-8555, Japan
| | - Rika Yasuhara
- Division of Pathology, Department of Oral Diagnostic Sciences, School of Dentistry, Showa University, Tokyo, 142-8555, Japan.
| | - Seya Kang
- Division of Dentistry for Persons with Disabilities, Department of Perioperative Medicine, School of Dentistry, Showa University, Tokyo, 142-8555, Japan
| | - Takahiro Funatsu
- Department of Pediatric Dentistry, School of Dentistry, Showa University, Tokyo, 142-8555, Japan
| | - Kenji Mishima
- Division of Pathology, Department of Oral Diagnostic Sciences, School of Dentistry, Showa University, Tokyo, 142-8555, Japan.
| |
Collapse
|
12
|
Engin AB, Engin A. MicroRNAs as Epigenetic Regulators of Obesity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:595-627. [PMID: 39287866 DOI: 10.1007/978-3-031-63657-8_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
In obesity, the process of adipogenesis largely determines the number of adipocytes in body fat depots. Adipogenesis is regulated by several adipocyte-selective micro-ribonucleic acids (miRNAs) and transcription factors that modulate adipocyte proliferation and differentiation. However, some miRNAs block the expression of master regulators of adipogenesis. Since the specific miRNAs display different expressions during adipogenesis, in mature adipocytes and permanent obesity, their use as biomarkers or therapeutic targets is feasible. Upregulated miRNAs in persistent obesity are downregulated during adipogenesis. Moreover, some of the downregulated miRNAs in obese individuals are upregulated in mature adipocytes. Induction of adipocyte stress and hypertrophy leads to the release of adipocyte-derived exosomes (AdEXs) that contain the cargo molecules, miRNAs. miRNAs are important messengers for intercellular communication involved in metabolic responses and have very specific signatures that direct the metabolic activity of target cells. While each miRNA targets multiple messenger RNAs (mRNAs), which may coordinate or antagonize each other's functions, several miRNAs are dysregulated in other tissues during obesity-related comorbidities. Deletion of the miRNA-processing enzyme DICER in pro-opiomelanocortin-expressing cells results in obesity, which is characterized by hyperphagia, increased adiposity, hyperleptinemia, defective glucose metabolism, and alterations in the pituitary-adrenal axis. In recent years, RNA-based therapeutical approaches have entered clinical trials as novel therapies against overweight and its complications. Development of lipid droplets, macrophage accumulation, macrophage polarization, tumor necrosis factor receptor-associated factor 6 activity, lipolysis, lipotoxicity, and insulin resistance are effectively controlled by miRNAs. Thereby, miRNAs as epigenetic regulators are used to determine the new gene transcripts and therapeutic targets.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Faculty of Pharmacy, Department of Toxicology, Gazi University, Hipodrom, Ankara, Turkey.
| | - Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey
| |
Collapse
|
13
|
Lyu M, Li F, Wang X, Xu K, Sun S. miR-145 Modulates Fatty Acid Metabolism by Targeting FOXO1 to Affect SERBP1 Activity in Bovine Mammary Epithelial Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:7440-7450. [PMID: 37154263 DOI: 10.1021/acs.jafc.2c09001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
MicroRNA-mediated gene regulation is important for the regulation of fatty acid metabolism and synthesis. Our previous study uncovered that the miR-145 expression is higher in the lactating mammary gland of dairy cows than in the dry-period, but the underlying molecular mechanism is incompletely understood. In this study, we have investigated the potential role of miR-145 in bovine mammary epithelial cells (BMECs). We found that the expression of miR-145 gradually increased during lactation. CRISPR/Cas9-mediated knockout (KO) of miR-145 in BMECs results in the downregulated expression of fatty acid metabolism-associated genes. Further results revealed that miR-145 KO reduced total triacylglycerol (TAG) and cholesterol (TC) accumulation and altered the composition of intracellular fatty acids (C16:0, C18:0, and C18:1). Conversely, miR-145 overexpression had the opposite effect. Bioinformatics online program predicted that miR-145 targets the 3'-UTR of the Forkhead Box O1 (FOXO1) gene. Subsequently, FOXO1 was identified as a direct target of miR-145 by qRT-PCR, Western blot analysis, and luciferase reporter assay. Furthermore, siRNA-mediated silencing of FOXO1 promoted fatty acid metabolism and TAG synthesis in BMECs. Additionally, we observed the involvement of FOXO1 in the transcriptional activity of the sterol regulatory element-binding protein 1 (SREBP1) gene promoter. Overall, our findings indicated that miR-145 relieves the inhibitory effect of FOXO1 on SREBP1 expression by targeting FOXO1 and subsequently regulating fatty acid metabolism. Thus, our results provide valuable information on the molecular mechanisms for improving milk yield and quality from the perspective of miRNA-mRNA networks.
Collapse
Affiliation(s)
- Ming Lyu
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, Xianyang Shaanxi 712100, PR China
| | - Fang Li
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, Xianyang Shaanxi 712100, PR China
| | - Xu Wang
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, Xianyang Shaanxi 712100, PR China
| | - Kun Xu
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling, Xianyang Shaanxi 712100, PR China
| | - Shuang Sun
- College of Life Science and Agricultural Engineering, Nanyang Normal University, Nanyang, Henan 473061, PR China
| |
Collapse
|
14
|
Cavati G, Pirrotta F, Merlotti D, Ceccarelli E, Calabrese M, Gennari L, Mingiano C. Role of Advanced Glycation End-Products and Oxidative Stress in Type-2-Diabetes-Induced Bone Fragility and Implications on Fracture Risk Stratification. Antioxidants (Basel) 2023; 12:antiox12040928. [PMID: 37107303 PMCID: PMC10135862 DOI: 10.3390/antiox12040928] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/06/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
Type 2 diabetes (T2D) and osteoporosis (OP) are major causes of morbidity and mortality that have arelevant health and economic burden. Recent epidemiological evidence suggests that both of these disorders are often associated with each other and that T2D patients have an increased risk of fracture, making bone an additional target of diabetes. As occurs for other diabetic complications, the increased accumulation of advanced glycation end-products (AGEs) and oxidative stress represent the major mechanisms explaining bone fragility in T2D. Both of these conditions directly and indirectly (through the promotion of microvascular complications) impair the structural ductility of bone and negatively affect bone turnover, leading to impaired bone quality, rather than decreased bone density. This makes diabetes-induced bone fragility remarkably different from other forms of OP and represents a major challenge for fracture risk stratification, since either the measurement of BMD or the use of common diagnostic algorithms for OP have a poor predictive value. We review and discuss the role of AGEs and oxidative stress on the pathophysiology of bone fragility in T2D, providing some indications on how to improve fracture risk prediction in T2D patients.
Collapse
Affiliation(s)
- Guido Cavati
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy
| | - Filippo Pirrotta
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy
| | - Daniela Merlotti
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy
| | - Elena Ceccarelli
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy
| | - Marco Calabrese
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy
| | - Luigi Gennari
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy
| | - Christian Mingiano
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy
| |
Collapse
|
15
|
Wang J, Zhang Y, Cao J, Wang Y, Anwar N, Zhang Z, Zhang D, Ma Y, Xiao Y, Xiao L, Wang X. The role of autophagy in bone metabolism and clinical significance. Autophagy 2023:1-19. [PMID: 36858962 PMCID: PMC10392742 DOI: 10.1080/15548627.2023.2186112] [Citation(s) in RCA: 124] [Impact Index Per Article: 62.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023] Open
Abstract
The skeletal system is the basis of the vertebral body composition, which affords stabilization sites for muscle attachment, protects vital organs, stores mineral ions, supplies places to the hematopoietic system, and participates in complex endocrine and immune system. Not surprisingly, bones are constantly reabsorbed, formed, and remodeled under physiological conditions. Once bone metabolic homeostasis is interrupted (including inflammation, tumors, fractures, and bone metabolic diseases), the body rapidly initiates bone regeneration to maintain bone tissue structure and quality. Macroautophagy/autophagy is an essential metabolic process in eukaryotic cells, which maintains metabolic energy homeostasis and plays a vital role in bone regeneration by controlling molecular degradation and organelle renewal. One relatively new observation is that mesenchymal cells, osteoblasts, osteoclasts, osteocytes, chondrocytes, and vascularization process exhibit autophagy, and the molecular mechanisms and targets involved are being explored and updated. The role of autophagy is also emerging in degenerative diseases (intervertebral disc degeneration [IVDD], osteoarthritis [OA], etc.) and bone metabolic diseases (osteoporosis [OP], osteitis deformans, osteosclerosis). The use of autophagy regulators to modulate autophagy has benefited bone regeneration, including MTOR (mechanistic target of rapamycin kinase) inhibitors, AMPK activators, and emerging phytochemicals. The application of biomaterials (especially nanomaterials) to trigger autophagy is also an attractive research direction, which can exert superior therapeutic properties from the material-loaded molecules/drugs or the material's properties such as shape, roughness, surface chemistry, etc. All of these have essential clinical significance with the discovery of autophagy associated signals, pathways, mechanisms, and treatments in bone diseases in the future.Abbreviations: Δψm: mitochondrial transmembrane potential AMPK: AMP-activated protein kinase ARO: autosomal recessive osteosclerosis ATF4: activating transcription factor 4 ATG: autophagy-related β-ECD: β-ecdysone BMSC: bone marrow mesenchymal stem cell ER: endoplasmic reticulum FOXO: forkhead box O GC: glucocorticoid HIF1A/HIF-1α: hypoxia inducible factor 1 subunit alpha HSC: hematopoietic stem cell HSP: heat shock protein IGF1: insulin like growth factor 1 IL1B/IL-1β: interleukin 1 beta IVDD: intervertebral disc degradation LPS: lipopolysaccharide MAPK: mitogen-activated protein kinase MSC: mesenchymal stem cell MTOR: mechanistic target of rapamycin kinase NP: nucleus pulposus NPWT: negative pressure wound therapy OA: osteoarthritis OP: osteoporosis PTH: parathyroid hormone ROS: reactive oxygen species SIRT1: sirtuin 1 SIRT3: sirtuin 3 SQSTM1/p62: sequestosome 1 TNFRSF11B/OPG: TNF receptor superfamily member 11b TNFRSF11A/RANK: tumor necrosis factor receptor superfamily, member 11a TNFSF11/RANKL: tumor necrosis factor (ligand) superfamily, member 11 TSC1: tuberous sclerosis complex 1 ULK1: unc-51 like autophagy activating kinase 1.
Collapse
Affiliation(s)
- Jing Wang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People's Republic of China
| | - Yi Zhang
- Department of Hygiene Toxicology, School of Public Health, Zunyi Medical University, Zunyi, Guizhou, People's Republic of China
| | - Jin Cao
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People's Republic of China
| | - Yi Wang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People's Republic of China
| | - Nadia Anwar
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People's Republic of China
| | - Zihan Zhang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People's Republic of China
| | - Dingmei Zhang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People's Republic of China
| | - Yaping Ma
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People's Republic of China
| | - Yin Xiao
- Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, Brisbane, Queensland, Australia.,School of Medicine and Dentistry & Menzies Health Institute Queensland, Griffith University, Queensland, Australia
| | - Lan Xiao
- School of Mechanical, Medical and Process Engineering, Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, Australia.,Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Xin Wang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, People's Republic of China.,School of Mechanical, Medical and Process Engineering, Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, Australia.,Australia-China Centre for Tissue Engineering and Regenerative Medicine, Queensland University of Technology, Brisbane, Queensland, Australia
| |
Collapse
|
16
|
De Sousa-Coelho AL, Gacias M, O'Neill BT, Relat J, Link W, Haro D, Marrero PF. FOXO1 represses PPARα-Mediated induction of FGF21 gene expression. Biochem Biophys Res Commun 2023; 644:122-129. [PMID: 36640666 DOI: 10.1016/j.bbrc.2023.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 01/05/2023] [Indexed: 01/07/2023]
Abstract
Fibroblast growth factor 21 (FGF21) has emerged as a metabolic regulator that exerts potent anti-diabetic and lipid-lowering effects in animal models of obesity and type 2 diabetes, showing a protective role in fatty liver disease and hepatocellular carcinoma progression. Hepatic expression of FGF21 is regulated by PPARα and is induced by fasting. Ablation of FoxO1 in liver has been shown to increase FGF21 expression in hyperglycemia. To better understand the role of FOXO1 in the regulation of FGF21 expression we have modified HepG2 human hepatoma cells to overexpress FoxO1 and PPARα. Here we show that FoxO1 represses PPARα-mediated FGF21 induction, and that the repression acts on the FGF21 gene promoter without affecting other PPARα target genes. Additionally, we demonstrate that FoxO1 physically interacts with PPARα and that FoxO1/3/4 depletion in skeletal muscle contributes to increased Fgf21 tissue levels. Taken together, these data indicate that FOXO1 is a PPARα-interacting protein that antagonizes PPARα activity on the FGF21 promoter. Because other PPARα target genes remained unaffected, these results suggest a highly specific mechanism implicated in FGF21 regulation. We conclude that FGF21 can be specifically modulated by FOXO1 in a PPARα-dependent manner.
Collapse
Affiliation(s)
- Ana Luísa De Sousa-Coelho
- Algarve Biomedical Center Research Institute (ABC-RI), Universidade do Algarve, Campus de Gambelas, Edifício 2, 8005-139, Faro, Portugal; Algarve Biomedical Center (ABC), Campus de Gambelas, Edifício 2, 8005-139, Faro, Portugal; Escola Superior de Saúde, Universidade do Algarve, Campus de Gambelas, Edifício 1, 8005-139, Faro, Portugal.
| | - Mar Gacias
- Department of Nutrition, Food Sciences and Gastronomy, School of Pharmacy and Food Sciences, Food Torribera Campus, University of Barcelona, E-08921, Santa Coloma de Gramenet, Spain
| | - Brian T O'Neill
- Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, 52242, Iowa, USA
| | - Joana Relat
- Department of Nutrition, Food Sciences and Gastronomy, School of Pharmacy and Food Sciences, Food Torribera Campus, University of Barcelona, E-08921, Santa Coloma de Gramenet, Spain; Institute of Nutrition and Food Safety of the University of Barcelona (INSA-UB), E-08921, Santa Coloma de Gramenet, Spain; CIBER Physiopathology of Obesity and Nutrition (CIBER-OBN), Instituto de Salud Carlos III, E-28029, Madrid, Spain
| | - Wolfgang Link
- Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), Arturo Duperier 4, 28029, Madrid, Spain
| | - Diego Haro
- Department of Nutrition, Food Sciences and Gastronomy, School of Pharmacy and Food Sciences, Food Torribera Campus, University of Barcelona, E-08921, Santa Coloma de Gramenet, Spain; CIBER Physiopathology of Obesity and Nutrition (CIBER-OBN), Instituto de Salud Carlos III, E-28029, Madrid, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), E-08028 Barcelona, Spain
| | - Pedro F Marrero
- Department of Nutrition, Food Sciences and Gastronomy, School of Pharmacy and Food Sciences, Food Torribera Campus, University of Barcelona, E-08921, Santa Coloma de Gramenet, Spain; CIBER Physiopathology of Obesity and Nutrition (CIBER-OBN), Instituto de Salud Carlos III, E-28029, Madrid, Spain; Institute of Biomedicine of the University of Barcelona (IBUB), E-08028 Barcelona, Spain.
| |
Collapse
|
17
|
Kandoor A, Fierst J. Dauer fate in a Caenorhabditis elegans Boolean network model. PeerJ 2023; 11:e14713. [PMID: 36710867 PMCID: PMC9879150 DOI: 10.7717/peerj.14713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 12/16/2022] [Indexed: 01/24/2023] Open
Abstract
Cellular fates are determined by genes interacting across large, complex biological networks. A critical question is how to identify causal relationships spanning distinct signaling pathways and underlying organismal phenotypes. Here, we address this question by constructing a Boolean model of a well-studied developmental network and analyzing information flows through the system. Depending on environmental signals Caenorhabditis elegans develop normally to sexual maturity or enter a reproductively delayed, developmentally quiescent 'dauer' state, progressing to maturity when the environment changes. The developmental network that starts with environmental signal and ends in the dauer/no dauer fate involves genes across 4 signaling pathways including cyclic GMP, Insulin/IGF-1, TGF-β and steroid hormone synthesis. We identified three stable motifs leading to normal development, each composed of genes interacting across the Insulin/IGF-1, TGF-β and steroid hormone synthesis pathways. Three genes known to influence dauer fate, daf-2, daf-7 and hsf-1, acted as driver nodes in the system. Using causal logic analysis, we identified a five gene cyclic subgraph integrating the information flow from environmental signal to dauer fate. Perturbation analysis showed that a multifactorial insulin profile determined the stable motifs the system entered and interacted with daf-12 as the switchpoint driving the dauer/no dauer fate. Our results show that complex organismal systems can be distilled into abstract representations that permit full characterization of the causal relationships driving developmental fates. Analyzing organismal systems from this perspective of logic and function has important implications for studies examining the evolution and conservation of signaling pathways.
Collapse
Affiliation(s)
- Alekhya Kandoor
- Biomedical Engineering, University of Virginia, Charlottesville, VA, United States of America
| | - Janna Fierst
- Biomolecular Sciences Institute and Department of Biology, Florida International University, Miami, FL, United States of America
| |
Collapse
|
18
|
Ramírez-Zamudio GD, Ganga MJG, Pereira GL, Nociti RP, Chiaratti MR, Cooke RF, Chardulo LAL, Baldassini WA, Machado-Neto OR, Curi RA. Effect of Cow-Calf Supplementation on Gene Expression, Processes, and Pathways Related to Adipogenesis and Lipogenesis in Longissimus thoracis Muscle of F1 Angus × Nellore Cattle at Weaning. Metabolites 2023; 13:metabo13020160. [PMID: 36837780 PMCID: PMC9962728 DOI: 10.3390/metabo13020160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/12/2023] [Accepted: 01/18/2023] [Indexed: 01/25/2023] Open
Abstract
The aim of this study was to identify differentially expressed genes, biological processes, and metabolic pathways related to adipogenesis and lipogenesis in calves receiving different diets during the cow-calf phase. Forty-eight uncastrated F1 Angus × Nellore males were randomly assigned to two treatments from thirty days of age to weaning: no creep feeding (G1) or creep feeding (G2). The creep feed offered contained ground corn (44.8%), soybean meal (40.4%), and mineral core (14.8%), with 22% crude protein and 65% total digestible nutrients in dry matter. After weaning, the animals were feedlot finished for 180 days and fed a single diet containing 12.6% forage and 87.4% corn-based concentrate. Longissimus thoracis muscle samples were collected by biopsy at weaning for transcriptome analysis and at slaughter for the measurement of intramuscular fat content (IMF) and marbling score (MS). Animals of G2 had 17.2% and 14.0% higher IMF and MS, respectively (p < 0.05). We identified 947 differentially expressed genes (log2 fold change 0.5, FDR 5%); of these, 504 were upregulated and 443 were downregulated in G2. Part of the genes upregulated in G2 were related to PPAR signaling (PPARA, SLC27A1, FABP3, and DBI), unsaturated fatty acid synthesis (FADS1, FADS2, SCD, and SCD5), and fatty acid metabolism (FASN, FADS1, FADS2, SCD, and SCD5). Regarding biological processes, the genes upregulated in G2 were related to cholesterol biosynthesis (EBP, CYP51A1, DHCR24, and LSS), unsaturated fatty acid biosynthesis (FADS2, SCD, SCD5, and FADS1), and insulin sensitivity (INSIG1 and LPIN2). Cow-calf supplementation G2 positively affected energy metabolism and lipid biosynthesis, and thus favored the deposition of marbling fat during the postweaning period, which was shown here in an unprecedented way, by analyzing the transcriptome, genes, pathways, and enriched processes due to the use of creep feeding.
Collapse
Affiliation(s)
| | - Maria Júlia Generoso Ganga
- School of Agriculture and Veterinary Sciences (FCAV), São Paulo State University (UNESP), Jaboticabal 14884-900, SP, Brazil
| | - Guilherme Luis Pereira
- School of Agriculture and Veterinary Sciences (FCAV), São Paulo State University (UNESP), Jaboticabal 14884-900, SP, Brazil
- School of Veterinary Medicine and Animal Science (FMVZ), São Paulo State University (UNESP), Botucatu 18618-681, SP, Brazil
| | - Ricardo Perecin Nociti
- College of Animal Science and Food Engineering, São Paulo University (USP), Pirassununga 13635-900, SP, Brazil
| | - Marcos Roberto Chiaratti
- Department of Genetics and Evolution, Federal University of São Carlos (UFSCAR), São Carlos 13565-905, SP, Brazil
| | | | - Luis Artur Loyola Chardulo
- School of Agriculture and Veterinary Sciences (FCAV), São Paulo State University (UNESP), Jaboticabal 14884-900, SP, Brazil
- School of Veterinary Medicine and Animal Science (FMVZ), São Paulo State University (UNESP), Botucatu 18618-681, SP, Brazil
| | - Welder Angelo Baldassini
- School of Agriculture and Veterinary Sciences (FCAV), São Paulo State University (UNESP), Jaboticabal 14884-900, SP, Brazil
- School of Veterinary Medicine and Animal Science (FMVZ), São Paulo State University (UNESP), Botucatu 18618-681, SP, Brazil
| | - Otávio Rodrigues Machado-Neto
- School of Agriculture and Veterinary Sciences (FCAV), São Paulo State University (UNESP), Jaboticabal 14884-900, SP, Brazil
- School of Veterinary Medicine and Animal Science (FMVZ), São Paulo State University (UNESP), Botucatu 18618-681, SP, Brazil
| | - Rogério Abdallah Curi
- School of Agriculture and Veterinary Sciences (FCAV), São Paulo State University (UNESP), Jaboticabal 14884-900, SP, Brazil
- School of Veterinary Medicine and Animal Science (FMVZ), São Paulo State University (UNESP), Botucatu 18618-681, SP, Brazil
- Correspondence:
| |
Collapse
|
19
|
Lee S, Usman TO, Yamauchi J, Chhetri G, Wang X, Coudriet GM, Zhu C, Gao J, McConnell R, Krantz K, Rajasundaram D, Singh S, Piganelli J, Ostrowska A, Soto-Gutierrez A, Monga SP, Singhi AD, Muzumdar RH, Tsung A, Dong HH. Myeloid FoxO1 depletion attenuates hepatic inflammation and prevents nonalcoholic steatohepatitis. J Clin Invest 2022; 132:154333. [PMID: 35700043 PMCID: PMC9282937 DOI: 10.1172/jci154333] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 05/27/2022] [Indexed: 11/17/2022] Open
Abstract
Hepatic inflammation is culpable for the evolution of asymptomatic steatosis to nonalcoholic steatohepatitis (NASH). Hepatic inflammation results from abnormal macrophage activation. We found that FoxO1 links overnutrition to hepatic inflammation by regulating macrophage polarization and activation. FoxO1 was upregulated in hepatic macrophages, correlating with hepatic inflammation, steatosis and fibrosis in mice and patients with NASH. Myeloid cell-conditional FoxO1 knockout skewed macrophage polarization from pro-inflammatory M1 to anti-inflammatory M2 phenotypes, accompanied by the reduction of macrophage infiltration in liver. These effects mitigated overnutrition-induced hepatic inflammation and insulin resistance, contributing to improved hepatic metabolism and increased energy expenditure in myeloid cell FoxO1 knockout mice on HFD. When fed a NASH-inducing diet, myeloid cell FoxO1 knockout mice were protected from developing NASH, culminating in the reduction of hepatic inflammation, steatosis and fibrosis. Mechanistically, FoxO1 counteracts Stat6 to skew macrophage polarization from M2 toward M1 signatures to perpetuate hepatic inflammation in NASH. FoxO1 appears as a pivotal mediator of macrophage activation in response to overnutrition and a therapeutic target for ameliorating hepatic inflammation to stem the disease progression from benign steatosis to NASH.
Collapse
Affiliation(s)
- Sojin Lee
- Department of Pediatrics, Children's Hospital, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Taofeek O Usman
- Department of Pediatrics, Children's Hospital, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Jun Yamauchi
- Department of Pediatrics, Children's Hospital, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Goma Chhetri
- Department of Pediatrics, Children's Hospital, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Xingchun Wang
- Department of Pediatrics, Children's Hospital, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Gina M Coudriet
- Department of Surgery, Children's Hospital, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Cuiling Zhu
- Department of Pediatrics, Children's Hospital, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Jingyang Gao
- Department of Pediatrics, Children's Hospital, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Riley McConnell
- Department of Pediatrics, Children's Hospital, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Kyler Krantz
- Department of Pediatrics, Children's Hospital, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Dhivyaa Rajasundaram
- Department of Pediatrics, Children's Hospital, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Sucha Singh
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Jon Piganelli
- Department of Pediatrics, Children's Hospital, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Alina Ostrowska
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Alejandro Soto-Gutierrez
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Satdarshan P Monga
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Aatur D Singhi
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Radhika H Muzumdar
- Department of Pediatrics, Children's Hospital, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| | - Allan Tsung
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, United States of America
| | - H Henry Dong
- Department of Pediatrics, Children's Hospital, University of Pittsburgh School of Medicine, Pittsburgh, United States of America
| |
Collapse
|
20
|
Huang CJ, Choo KB, Chen CF. The MicroRNA-Signaling-Peroxisome Proliferator-Activated Receptor Gamma Connection in the Modulation of Adipogenesis: Bioinformatics Projection on Chicken. Poult Sci 2022; 101:101950. [PMID: 35689996 PMCID: PMC9192975 DOI: 10.1016/j.psj.2022.101950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 03/19/2022] [Accepted: 04/15/2022] [Indexed: 10/29/2022] Open
|
21
|
Nathanael J, Suardana P, Vianney YM, Dwi Putra SE. The role of FoxO1 and its modulation with small molecules in the development of diabetes mellitus: A review. Chem Biol Drug Des 2021; 99:344-361. [PMID: 34862852 DOI: 10.1111/cbdd.13989] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 11/11/2021] [Accepted: 11/21/2021] [Indexed: 12/14/2022]
Abstract
Diabetes mellitus type 2 (T2D) is one of the metabolic disorders suffered by a global human being. Certain factors, such as lifestyle and heredity, can increase a person's tendency for T2D. Various genes and proteins play a role in the development of insulin resistance and ultimately diabetes in which one central protein that is discussed in this review is FoxO1. In this review, we regard FoxO1 activation as detrimental, promote high plasma glucose level, and induce insulin resistance. Indeed, many contrasting studies arise since FoxO1 is an important protein to alleviate oxidative stress and promote cell survival, for example, also by preventing hyperglycemic-induced cell death. Inter-relation to PPARG, another important protein in metabolism, is also discussed. Ultimately, we discussed contrasting approaches of targeting FoxO1 to combat diabetes mellitus by small molecules.
Collapse
Affiliation(s)
- Joshua Nathanael
- Department of Biotechnology, Faculty of Biotechnology, University of Surabaya, Surabaya, East Java, Indonesia
| | - Putu Suardana
- Department of Biotechnology, Faculty of Biotechnology, University of Surabaya, Surabaya, East Java, Indonesia
| | - Yoanes Maria Vianney
- Department of Biotechnology, Faculty of Biotechnology, University of Surabaya, Surabaya, East Java, Indonesia
| | - Sulistyo Emantoko Dwi Putra
- Department of Biotechnology, Faculty of Biotechnology, University of Surabaya, Surabaya, East Java, Indonesia
| |
Collapse
|
22
|
Abstract
The intimate association between obesity and type II diabetes urges for a deeper understanding of adipocyte function. We and others have previously delineated a role for the tumor suppressor p53 in adipocyte biology. Here, we show that mice haploinsufficient for MDM2, a key regulator of p53, in their adipose stores suffer from overt obesity, glucose intolerance, and hepatic steatosis. These mice had decreased levels of circulating palmitoleic acid [non-esterified fatty acid (NEFA) 16:1] concomitant with impaired visceral adipose tissue expression of Scd1 and Ffar4. A similar decrease in Scd and Ffar4 expression was found in in vitro differentiated adipocytes with perturbed MDM2 expression. Lowered MDM2 levels led to nuclear exclusion of the transcriptional cofactors, MORC2 and LIPIN1, and thereby possibly hampered adipocyte function by antagonizing LIPIN1-mediated PPARγ coactivation. Collectively, these data argue for a hitherto unknown interplay between MDM2 and MORC2/LIPIN1 involved in balancing adipocyte function.
Collapse
|
23
|
Guru B, Tamrakar AK, Mandal SP, Kumar PBR, Sharma A, Manjula SN. A Novel Partial PPARγ Agonist Has Weaker Lipogenic Effect in Adipocytes and Stimulates GLUT4 Translocation in Skeletal Muscle Cells via AMPK-Dependent Signaling. Pharmacology 2021; 107:90-101. [PMID: 34736259 DOI: 10.1159/000519331] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 08/29/2021] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Peroxisome proliferator-activated receptor gamma (PPARγ) agonists are highly effective in treating insulin resistance. However, associated side effects such as weight gain due to increase in adipogenesis and lipogenesis hinder their clinical use. The aim of the study was to design and synthesize novel partial PPARγ agonists with weaker lipogenic effect in adipocytes and enhanced glucose transporter 4 (GLUT4) translocation stimulatory effect in skeletal muscle cells. METHODS Novel partial PPARγ agonists (GS1, GS2, and GS3) were designed and screened to predict their binding interactions with PPARγ by molecular docking. The stability of the docked ligand-PPARγ complex was studied by molecular dynamics (MD) simulation. The cytotoxicity of synthesized compounds was tested in 3T3-L1 adipocytes and L6 myoblasts by MTT assay. The lipogenic effect was investigated in 3T3-L1 adipocytes using oil red O staining and GLUT4 translocation stimulatory effect in L6-GLUT4myc myotubes by an antibody-coupled colorimetric assay. RESULTS The molecular docking showed the binding interactions between designed agonists and PPARγ. MD simulation demonstrated good stability between the GS2-PPARγ complex. GS2 and GS3 did not show any significant effect on cell viability up to 80 or 100 μM concentration. Pioglitazone treatment significantly increased intracellular lipid accumulation in adipocytes compared to control. However, this effect was significantly less in GS2- and GS3-treated conditions compared to pioglitazone at 10 μM concentration, indicating weaker lipogenic effect. Furthermore, GS2 significantly stimulated GLUT4 translocation to the plasma membrane in a dose-dependent manner via the AMPK-dependent signaling pathway in skeletal muscle cells. CONCLUSION GS2 may be a promising therapeutic agent for the treatment of insulin resistance and type 2 diabetes mellitus without adiposity.
Collapse
Affiliation(s)
- Bhavimani Guru
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
| | - Akhilesh K Tamrakar
- Division of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Subhankar P Mandal
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
| | - Prashantha B R Kumar
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
| | - Aditya Sharma
- Division of Biochemistry and Structural Biology, CSIR-Central Drug Research Institute, Lucknow, India
| | | |
Collapse
|
24
|
Tebbe AW, Hanson J, Weiss WP. Effects of metabolizable protein concentration, amino acid profile, and fiber source on the messenger RNA expression of skeletal muscle in peripartum dairy cows. J Dairy Sci 2021; 104:7888-7901. [PMID: 33814155 DOI: 10.3168/jds.2021-20176] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 02/25/2021] [Indexed: 01/11/2023]
Abstract
After parturition, dairy cows mobilize AA from skeletal muscle to meet metabolizable protein (MP) requirements. High mobilization may compromise cow health and longer-term milk production. Postpartum diets with higher MP concentrations, improved AA profiles, or MP increased at the expense of forages rather than nonforage fiber sources may attenuate muscle catabolism; however, the molecular mechanisms responsible need investigation. We evaluated mRNA expression in the longissimus dorsi of cows fed postpartum diets differing in MP concentration, AA profile, and fiber source. From 0 to 25 d after parturition, 40 multiparous cows received the following diets: (1) 13% deficient in MP (D-MP), (2) adequate in MP using primarily soy protein (A-MP), (3) adequate in MP using blends of proteins and individual AA to improve the AA profile (Blend), or (4) similar to Blend except additional protein replaced forage (Blend-fNDF). Biopsies were taken approximately -5, 7, and 25 d relative to parturition. Greater dietary MP concentration (D-MP vs. A-MP and Blend) decreased expression of genes related to protein synthesis (MTOR, RPS6KB1) and degradation (FOXO1), inflammation (IFNG, TLR4), and endoplasmic reticulum (ER) stress (HSPA5, DDIT) and increased genes associated with lipogenesis (PPARG) and glucose oxidation (LDH, MB). In Blend versus A-MP (i.e., effect of AA profile), expression related to apoptosis (CASP8) and inflammation (TNFA) decreased and genes associated with cell cycle progression (E2F1) and fast-twitch glycolytic muscle fiber type (MYH4) increased. Less forage (Blend-fNDF vs. Blend) decreased genes associated with lipogenesis (PPARG, ACACA) and ER stress (BCL2, DDIT3, EIF2AK3, PPP1R15A) and increased genes associated with inflammation (TNF), inhibition of myogenesis (MSTN), and autophagy (PEBP1). In summary and based on mRNA expression, increasing MP supply may attenuate muscle turnover and ER stress. However, an unbalanced AA supply reduced cell cycle progression and protein synthesis. Lower energy supplies may reduce cell growth and cause autophagy.
Collapse
Affiliation(s)
- Alexander W Tebbe
- Department of Animal Sciences, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster 44691
| | - Juliette Hanson
- Food Animal Health Research Program, The Ohio State University, Wooster 44691
| | - William P Weiss
- Department of Animal Sciences, Ohio Agricultural Research and Development Center, The Ohio State University, Wooster 44691.
| |
Collapse
|
25
|
Kim S, Lee N, Park ES, Yun H, Ha TU, Jeon H, Yu J, Choi S, Shin B, Yu J, Rhee SD, Choi Y, Rho J. T-Cell Death Associated Gene 51 Is a Novel Negative Regulator of PPARγ That Inhibits PPARγ-RXRα Heterodimer Formation in Adipogenesis. Mol Cells 2021; 44:1-12. [PMID: 33335079 PMCID: PMC7854182 DOI: 10.14348/molcells.2020.0143] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 11/06/2020] [Accepted: 11/27/2020] [Indexed: 11/27/2022] Open
Abstract
The nuclear receptor peroxisome proliferator-activated receptor γ (PPARγ) is the master transcriptional regulator in adipogenesis. PPARγ forms a heterodimer with another nuclear receptor, retinoid X receptor (RXR), to form an active transcriptional complex, and their transcriptional activity is tightly regulated by the association with either coactivators or corepressors. In this study, we identified T-cell death-associated gene 51 (TDAG51) as a novel corepressor of PPARγ-mediated transcriptional regulation. We showed that TDAG51 expression is abundantly maintained in the early stage of adipogenic differentiation. Forced expression of TDAG51 inhibited adipocyte differentiation in 3T3-L1 cells. We found that TDAG51 physically interacts with PPARγ in a ligand-independent manner. In deletion mutant analyses, large portions of the TDAG51 domains, including the pleckstrin homology-like, glutamine repeat and proline-glutamine repeat domains but not the proline-histidine repeat domain, are involved in the interaction with the region between residues 140 and 506, including the DNA binding domain, hinge, ligand binding domain and activation function-2 domain, in PPARγ. The heterodimer formation of PPARγ-RXRα was competitively inhibited in a ligand-independent manner by TDAG51 binding to PPARγ. Thus, our data suggest that TDAG51, which could determine adipogenic cell fate, acts as a novel negative regulator of PPARγ by blocking RXRα recruitment to the PPARγ-RXRα heterodimer complex in adipogenesis.
Collapse
Affiliation(s)
- Sumi Kim
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon 34134, Korea
| | - Nari Lee
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon 34134, Korea
| | - Eui-Soon Park
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon 34134, Korea
| | - Hyeongseok Yun
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon 34134, Korea
| | - Tae-Uk Ha
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon 34134, Korea
| | - Hyoeun Jeon
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon 34134, Korea
| | - Jiyeon Yu
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon 34134, Korea
| | - Seunga Choi
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon 34134, Korea
| | - Bongjin Shin
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon 34134, Korea
| | - Jungeun Yu
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon 34134, Korea
| | - Sang Dal Rhee
- Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon 34114, Korea
| | - Yongwon Choi
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Jaerang Rho
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon 34134, Korea
| |
Collapse
|
26
|
Zhao N, Tan H, Wang L, Han L, Cheng Y, Feng Y, Li T, Liu X. Palmitate induces fat accumulation via repressing FoxO1-mediated ATGL-dependent lipolysis in HepG2 hepatocytes. PLoS One 2021; 16:e0243938. [PMID: 33449950 PMCID: PMC7810308 DOI: 10.1371/journal.pone.0243938] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 11/30/2020] [Indexed: 02/05/2023] Open
Abstract
Obesity is closely associated with non-alcoholic fatty liver disease (NAFLD), and elevated serum palmitate is the link between obesity and excessive hepatic lipid accumulation. Forkhead box O-1 (FoxO1) is one of the FoxO family members of transcription factors and can stimulate adipose triglyceride lipase (ATGL) and suppress its inhibitor G0/G1 switch gene 2 (G0S2) expression in the liver. However, previous researches have also shown conflicting results regarding the role of FoxO1 in hepatic lipid accumulation. We therefore examined the role of FoxO1 as a downstream suppressor to palmitate-stimulated hepatic steatosis. Palmitate significantly promoted lipid accumulation but inhibited lipid decomposition in human HepG2 hepatoma cells. Palmitate also significantly reduced FoxO1, ATGL and its activator comparative gene identification-58 (CGI-58) expression but increased peroxisome proliferator-activated receptorγ (PPARγ) and its target gene G0S2 expression. FoxO1 overexpression significantly increased palmitate-inhibited ATGL and CGI-58 expression but reduced palmitate-stimulated PPARγ and its target gene G0S2 expression. FoxO1 overexpression also inhibited lipid accumulation and promoted lipolysis in palmitate-treated hepatocytes. Overall, these results indicate that FoxO1-mediated ATGL-dependent lipolysis may be an effective molecular mechanism in protecting hepatocytes from palmitate-induced fat accumulation.
Collapse
Affiliation(s)
- Naiqian Zhao
- Department of Gerontology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- * E-mail:
| | - Huiwen Tan
- Department of Endocrinology and Metabolism, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Li Wang
- Department of Gerontology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Le Han
- Department of Gerontology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yanli Cheng
- Department of Gerontology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Ying Feng
- Department of Gerontology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Ting Li
- Department of Gerontology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiaoling Liu
- Department of Gerontology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
27
|
Calissi G, Lam EWF, Link W. Therapeutic strategies targeting FOXO transcription factors. Nat Rev Drug Discov 2021; 20:21-38. [PMID: 33173189 DOI: 10.1038/s41573-020-0088-2] [Citation(s) in RCA: 209] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/28/2020] [Indexed: 12/13/2022]
Abstract
FOXO proteins are transcription factors that are involved in numerous physiological processes and in various pathological conditions, including cardiovascular disease, cancer, diabetes and chronic neurological diseases. For example, FOXO proteins are context-dependent tumour suppressors that are frequently inactivated in human cancers, and FOXO3 is the second most replicated gene associated with extreme human longevity. Therefore, pharmacological manipulation of FOXO proteins is a promising approach to developing therapeutics for cancer and for healthy ageing. In this Review, we overview the role of FOXO proteins in health and disease and discuss the pharmacological approaches to modulate FOXO function.
Collapse
Affiliation(s)
- Giampaolo Calissi
- Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), Madrid, Spain
| | - Eric W-F Lam
- Department of Surgery and Cancer, Imperial College London, Imperial Centre for Translational and Experimental Medicine (ICTEM), London, UK
| | - Wolfgang Link
- Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), Madrid, Spain.
| |
Collapse
|
28
|
Sun J, Lou Y, Zhu J, Shen H, Zhou D, Zhu L, Yang X, Xie M, Li L, Huang X, Zhu M, Zheng Y, Xie W, Ye X, Jin J, Zhu HH. Hypertriglyceridemia in Newly Diagnosed Acute Promyelocytic Leukemia. Front Oncol 2020; 10:577796. [PMID: 33324553 PMCID: PMC7724081 DOI: 10.3389/fonc.2020.577796] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/26/2020] [Indexed: 01/12/2023] Open
Abstract
The primary aim of the present retrospective study was to investigate lipid profiles and kinetics in acute promyelocytic leukemia (APL) patients. We analyzed 402 newly diagnosed APL patients and 201 non-APL patients with acute myeloid leukemia (as control). Incidence of hypertriglyceridemia in APL patients and non-APL patients was 55.82% and 28.4% (p = 0.0003). The initial levels of triglycerides, total cholesterol, high-density lipoprotein cholesterol and low-density lipoprotein cholesterol were higher in APL patients than in control (all p < 0.0001). In APL patients, triglyceride levels were significantly increased during induction treatment with all-trans retinoic acid and arsenic. Multivariable analysis showed that age, being overweight (body mass index ≥25) and APL were independent risk factors for hypertriglyceridemia in all patients before treatment. High triglyceride levels were not significantly associated with disease-free survival or overall survival in the APL patients. In summary, in the current study triglyceride levels were significantly elevated in APL patients before treatment, and they increased during induction treatment, but there were no significant corresponding effects on survival.
Collapse
Affiliation(s)
- Jianai Sun
- Department of Hematology, The First Affiliated Hospital of Medical School of Zhejiang University, Hangzhou, Zhejiang, China.,Zhejiang Province Key Laboratory of Hematology Oncology Diagnosis and Treatment, Hangzhou, China
| | - Yinjun Lou
- Department of Hematology, The First Affiliated Hospital of Medical School of Zhejiang University, Hangzhou, Zhejiang, China.,Zhejiang Province Key Laboratory of Hematology Oncology Diagnosis and Treatment, Hangzhou, China
| | - Jingjing Zhu
- Department of Hematology, The First Affiliated Hospital of Medical School of Zhejiang University, Hangzhou, Zhejiang, China.,Zhejiang Province Key Laboratory of Hematology Oncology Diagnosis and Treatment, Hangzhou, China
| | - Huafei Shen
- Department of Hematology, The First Affiliated Hospital of Medical School of Zhejiang University, Hangzhou, Zhejiang, China.,Zhejiang Province Key Laboratory of Hematology Oncology Diagnosis and Treatment, Hangzhou, China
| | - De Zhou
- Department of Hematology, The First Affiliated Hospital of Medical School of Zhejiang University, Hangzhou, Zhejiang, China.,Zhejiang Province Key Laboratory of Hematology Oncology Diagnosis and Treatment, Hangzhou, China
| | - Lixia Zhu
- Department of Hematology, The First Affiliated Hospital of Medical School of Zhejiang University, Hangzhou, Zhejiang, China.,Zhejiang Province Key Laboratory of Hematology Oncology Diagnosis and Treatment, Hangzhou, China
| | - Xiudi Yang
- Department of Hematology, The First Affiliated Hospital of Medical School of Zhejiang University, Hangzhou, Zhejiang, China.,Zhejiang Province Key Laboratory of Hematology Oncology Diagnosis and Treatment, Hangzhou, China
| | - Mixue Xie
- Department of Hematology, The First Affiliated Hospital of Medical School of Zhejiang University, Hangzhou, Zhejiang, China.,Zhejiang Province Key Laboratory of Hematology Oncology Diagnosis and Treatment, Hangzhou, China
| | - Li Li
- Department of Hematology, The First Affiliated Hospital of Medical School of Zhejiang University, Hangzhou, Zhejiang, China.,Zhejiang Province Key Laboratory of Hematology Oncology Diagnosis and Treatment, Hangzhou, China
| | - Xianbo Huang
- Department of Hematology, The First Affiliated Hospital of Medical School of Zhejiang University, Hangzhou, Zhejiang, China.,Zhejiang Province Key Laboratory of Hematology Oncology Diagnosis and Treatment, Hangzhou, China
| | - Mingyu Zhu
- Department of Hematology, The First Affiliated Hospital of Medical School of Zhejiang University, Hangzhou, Zhejiang, China.,Zhejiang Province Key Laboratory of Hematology Oncology Diagnosis and Treatment, Hangzhou, China
| | - Yanlong Zheng
- Department of Hematology, The First Affiliated Hospital of Medical School of Zhejiang University, Hangzhou, Zhejiang, China.,Zhejiang Province Key Laboratory of Hematology Oncology Diagnosis and Treatment, Hangzhou, China
| | - Wanzhuo Xie
- Department of Hematology, The First Affiliated Hospital of Medical School of Zhejiang University, Hangzhou, Zhejiang, China.,Zhejiang Province Key Laboratory of Hematology Oncology Diagnosis and Treatment, Hangzhou, China
| | - Xiujin Ye
- Department of Hematology, The First Affiliated Hospital of Medical School of Zhejiang University, Hangzhou, Zhejiang, China.,Zhejiang Province Key Laboratory of Hematology Oncology Diagnosis and Treatment, Hangzhou, China
| | - Jie Jin
- Department of Hematology, The First Affiliated Hospital of Medical School of Zhejiang University, Hangzhou, Zhejiang, China.,Zhejiang Province Key Laboratory of Hematology Oncology Diagnosis and Treatment, Hangzhou, China
| | - Hong-Hu Zhu
- Department of Hematology, The First Affiliated Hospital of Medical School of Zhejiang University, Hangzhou, Zhejiang, China.,Zhejiang Province Key Laboratory of Hematology Oncology Diagnosis and Treatment, Hangzhou, China
| |
Collapse
|
29
|
Peroxisome Proliferator-Activated Receptors as Molecular Links between Caloric Restriction and Circadian Rhythm. Nutrients 2020; 12:nu12113476. [PMID: 33198317 PMCID: PMC7696073 DOI: 10.3390/nu12113476] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 11/04/2020] [Accepted: 11/09/2020] [Indexed: 02/06/2023] Open
Abstract
The circadian rhythm plays a chief role in the adaptation of all bodily processes to internal and environmental changes on the daily basis. Next to light/dark phases, feeding patterns constitute the most essential element entraining daily oscillations, and therefore, timely and appropriate restrictive diets have a great capacity to restore the circadian rhythm. One of the restrictive nutritional approaches, caloric restriction (CR) achieves stunning results in extending health span and life span via coordinated changes in multiple biological functions from the molecular, cellular, to the whole-body levels. The main molecular pathways affected by CR include mTOR, insulin signaling, AMPK, and sirtuins. Members of the family of nuclear receptors, the three peroxisome proliferator-activated receptors (PPARs), PPARα, PPARβ/δ, and PPARγ take part in the modulation of these pathways. In this non-systematic review, we describe the molecular interconnection between circadian rhythm, CR-associated pathways, and PPARs. Further, we identify a link between circadian rhythm and the outcomes of CR on the whole-body level including oxidative stress, inflammation, and aging. Since PPARs contribute to many changes triggered by CR, we discuss the potential involvement of PPARs in bridging CR and circadian rhythm.
Collapse
|
30
|
Zhu Y, Bai J, Zhou Y, Zhang Y, Zhao Y, Dong Y, Xiao X. Water-soluble and alkali-soluble polysaccharides from bitter melon inhibited lipid accumulation in HepG2 cells and Caenorhabditis elegans. Int J Biol Macromol 2020; 166:155-165. [PMID: 33172614 DOI: 10.1016/j.ijbiomac.2020.10.128] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 10/12/2020] [Accepted: 10/15/2020] [Indexed: 12/19/2022]
Abstract
Bitter melon polysaccharides (BPS) have been reported to have hypolipidemic effects. However, the precise mechanism of BPS regulating lipid metabolism remains elusive. Water-soluble (WBPS) and alkali-soluble bitter melon polysaccharides (ABPS) were extracted to evaluate the fat-lowering bioactivities in HepG2 cells and Caenorhabditis elegans. WBPS and ABPS were slightly different in the uronic acid contents (22.23% and 5.69%), monosaccharide composition, molecular weight (Mw: 332 kDa and 1552 kDa, respectively) and IR spectra. In palmitic acid-treated HepG2 cell, the ABPS exhibited better effects on accelerating glucose consumption and decreasing the triglyceride content than WBPS via stimulating glucose consumption (GLUT4) and gluconeogenesis (PEPCK). In the model of glucose-treated C. elegans, we observed that both WBPS and ABPS obviously suppressed the fat accumulation, more significantly by ABPS, along with no toxicity towards some physical activities. Fat-5, fat-6 and fat-7 mediated fatty acid desaturases pathways were further confirmed to be involved in the lipid-lowering effects of BPSs. Our studies demonstrated that both WBPS and ABPS can exhibit effects on fat- lowering in HepG2 cells and C. elegans.
Collapse
Affiliation(s)
- Ying Zhu
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, PR China
| | - Juan Bai
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, PR China
| | - Yurong Zhou
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, PR China
| | - Yi Zhang
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, PR China
| | - Yansheng Zhao
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, PR China
| | - Ying Dong
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, PR China
| | - Xiang Xiao
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, PR China.
| |
Collapse
|
31
|
Farias-Pereira R, Savarese J, Yue Y, Lee SH, Park Y. Fat-lowering effects of isorhamnetin are via NHR-49-dependent pathway in Caenorhabditis elegans. Curr Res Food Sci 2020; 2:70-76. [PMID: 32914113 PMCID: PMC7473354 DOI: 10.1016/j.crfs.2019.11.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Isorhamnetin (3-O-methylquercetin), a flavonol found in dill weed, sea buckthorn berries, kale and onions, has been suggested to have anti-obesity effects, but there is limited evidence of its mechanisms of action on lipid metabolism. The goal of this study was to investigate the effects of isorhamnetin on lipid metabolism using Caenorhabditis elegans as an animal model. Isorhamnetin reduced fat accumulation without affecting food intake or energy expenditure in C. elegans. The isorhamnetin's fat-lowering effects were dependent on nhr-49, a homolog of the human peroxisome proliferator-activated receptor alpha (PPARα). Isorhamnetin upregulated an enoyl-CoA hydratase (ech-1.1, involved in fatty acid β-oxidation) and adipose triglyceride lipase (atgl-1, involved in lipolysis) via NHR-49-dependent pathway at transcriptional levels. Isorhamnetin also upregulated the C. elegans AMP-activated protein kinase (AMPK) subunits homologs (aak-1 and aak-2), involved in energy homeostasis. These results suggest that isorhamnetin reduces body fat by increasing fat oxidation in part via NHR-49/PPARα-dependent pathway. Isorhamnetin reduced fat accumulation in Caenorhabditis elegans. Food intake and energy expenditure were not changed by isorhamnetin. Isorhamnetin's fat-lowering effects were dependent on nhr-49/PPARα. Isorhamnetin upregulated transcriptionally AAK/AMPK, which may activate NHR-49. Isorhamnetin increased fat breakdown by upregulating ech-1.1/HADHA and atgl-1/ATGL.
Collapse
Affiliation(s)
| | - Jessica Savarese
- Department of Food Science, University of Massachusetts, Amherst, MA, 01003, USA
| | - Yiren Yue
- Department of Food Science, University of Massachusetts, Amherst, MA, 01003, USA
| | - Seong-Ho Lee
- Department of Nutrition and Food Science, University of Maryland, College Park, MD, 20742, USA
| | - Yeonhwa Park
- Department of Food Science, University of Massachusetts, Amherst, MA, 01003, USA
| |
Collapse
|
32
|
Li YZ, Di Cristofano A, Woo M. Metabolic Role of PTEN in Insulin Signaling and Resistance. Cold Spring Harb Perspect Med 2020; 10:a036137. [PMID: 31964643 PMCID: PMC7397839 DOI: 10.1101/cshperspect.a036137] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Phosphatase and tensin homolog (PTEN) is most prominently known for its function in tumorigenesis. However, a metabolic role of PTEN is emerging as a result of its altered expression in type 2 diabetes (T2D), which results in impaired insulin signaling and promotion of insulin resistance during the pathogenesis of T2D. PTEN functions in regulating insulin signaling across different organs have been identified. Through the use of a variety of models, such as tissue-specific knockout (KO) mice and in vitro cell cultures, PTEN's role in regulating insulin action has been elucidated across many cell types. Herein, we will review the recent advancements in the understanding of PTEN's metabolic functions in each of the tissues and cell types that contribute to regulating systemic insulin sensitivity and discuss how PTEN may represent a promising therapeutic strategy for treatment or prevention of T2D.
Collapse
Affiliation(s)
- Yu Zhe Li
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario M5G 2C4, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario M5G 2M9, Canada
| | - Antonio Di Cristofano
- Department of Developmental and Molecular Biology and Medicine (Oncology), Albert Einstein College of Medicine and Albert Einstein Cancer Center, Bronx, New York 10461, USA
| | - Minna Woo
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario M5G 2C4, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario M5G 2M9, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario M5G 2M9, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, University Health Network/Mount Sinai Hospital, University of Toronto, Toronto, Ontario M5G 2C4, Canada
| |
Collapse
|
33
|
Kim DH, Ha S, Choi YJ, Dong HH, Yu BP, Chung HY. Altered FoxO1 and PPARγ interaction in age-related ER stress-induced hepatic steatosis. Aging (Albany NY) 2020; 11:4125-4144. [PMID: 31246177 PMCID: PMC6628996 DOI: 10.18632/aging.102042] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 06/17/2019] [Indexed: 01/05/2023]
Abstract
Chronic kidney disease (CKD) is one of the most powerful predictors of premature cardiovascular disease (CVD), with heightened susceptibility to vascular intimal and medial calcification associated with a high cardiovascular mortality. Abnormal mineral metabolism of calcium (Ca) and phosphate (P) and underlying (dys)regulated hormonal control in CKD-mineral and bone disorder (MBD) is often accompanied by bone loss and increased vascular calcification (VC). While VC is known to be a multifactorial process and a major risk factor for CVD, the view of primary triggers and molecular mechanisms complexity has been shifting with novel scientific knowledge over the last years. In this review we highlight the importance of calcium-phosphate (CaP) mineral crystals in VC with an integrated view over the complexity of CKD, while discuss past and recent literature aiming to highlight novel horizons on this major health burden. Exacerbated VC in CKD patients might result from several interconnected mechanisms involving abnormal mineral metabolism, dysregulation of endogenous calcification inhibitors and inflammatory pathways, which function in a feedback loop driving disease progression and cardiovascular outcomes. We propose that novel approaches targeting simultaneously VC and inflammation might represent valuable new prognostic tools and targets for therapeutics and management of cardiovascular risk in the CKD population.
Collapse
Affiliation(s)
- Dae Hyun Kim
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 46241, Korea
| | - Sugyeong Ha
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 46241, Korea
| | - Yeon Ja Choi
- Department of Biopharmaceutical Engineering, Division of Chemistry and Biotechnology, Dongguk University, Gyeongju 38066, Korea
| | - H Henry Dong
- Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Byung Pal Yu
- Department of Physiology, The University of Texas Health Science Center at San Antonio, TX 78229, USA
| | - Hae Young Chung
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan 46241, Korea
| |
Collapse
|
34
|
Peroxisome Proliferator-Activated Receptors and Caloric Restriction-Common Pathways Affecting Metabolism, Health, and Longevity. Cells 2020; 9:cells9071708. [PMID: 32708786 PMCID: PMC7407644 DOI: 10.3390/cells9071708] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/14/2020] [Accepted: 07/14/2020] [Indexed: 02/06/2023] Open
Abstract
Caloric restriction (CR) is a traditional but scientifically verified approach to promoting health and increasing lifespan. CR exerts its effects through multiple molecular pathways that trigger major metabolic adaptations. It influences key nutrient and energy-sensing pathways including mammalian target of rapamycin, Sirtuin 1, AMP-activated protein kinase, and insulin signaling, ultimately resulting in reductions in basic metabolic rate, inflammation, and oxidative stress, as well as increased autophagy and mitochondrial efficiency. CR shares multiple overlapping pathways with peroxisome proliferator-activated receptors (PPARs), particularly in energy metabolism and inflammation. Consequently, several lines of evidence suggest that PPARs might be indispensable for beneficial outcomes related to CR. In this review, we present the available evidence for the interconnection between CR and PPARs, highlighting their shared pathways and analyzing their interaction. We also discuss the possible contributions of PPARs to the effects of CR on whole organism outcomes.
Collapse
|
35
|
Amyloid-beta (Aβ 1-42)-induced paralysis in Caenorhabditis elegans is reduced through NHR-49/PPARalpha. Neurosci Lett 2020; 730:135042. [PMID: 32413539 DOI: 10.1016/j.neulet.2020.135042] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 05/07/2020] [Indexed: 01/03/2023]
Abstract
Alzheimer´s disease is a neurodegenerative disorder characterized by the misfolding and aggregation of amyloid β (Aβ). Agonists of peroxisomal proliferator-activated receptors (PPARs) are discussed as anti-amyloidogenic compounds, e.g. due to their cholesterol-lowering activities. In a previous study we have shown in Caenorhabditis elegans expressing human Aβ in muscle cells, that inhibition of steroid-signaling, by RNAi of respective members of the signaling pathway or by reducing cellular cholesterol uptake, both increases the nuclear translocation of the foxo transcription factor DAF-16 and concomitantly reduces Aβ-induced paralysis. Using RNAi in the present study we show that NHR-49/PPARalpha inhibits steroidal-signaling upstream of DAF-9, a cytochrome P450-dependent enzyme which generates dafachronic acids as ligands for the nuclear hormone receptor DAF-12, and upstream of DAF-12 itself. The NHR-49/PPARalpha agonist fenofibrate reduces Aβ-induced paralysis in dependence on nhr-49 and nuclear translocation of DAF-16. In conclusion, activation of NHR-49/PPARalpha inhibits the steroidal-signaling pathway which increases the nuclear translocation of DAF-16 and inhibits the Aβ-induced phenotype in an Alzheimer model of C. elegans.
Collapse
|
36
|
Feng X, Zhu C, Lee S, Gao J, Zhu P, Yamauchi J, Pan C, Singh S, Qu S, Miller R, Monga SP, Peng Y, Dong HH. Depletion of hepatic forkhead box O1 does not affect cholelithiasis in male and female mice. J Biol Chem 2020; 295:7003-7017. [PMID: 32273342 DOI: 10.1074/jbc.ra119.012272] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 04/07/2020] [Indexed: 11/06/2022] Open
Abstract
Cholelithiasis is one of the most prevalent gastroenterological diseases and is characterized by the formation of gallstones in the gallbladder. Both clinical and preclinical data indicate that obesity, along with comorbidity insulin resistance, is a predisposing factor for cholelithiasis. Forkhead box O1 (FoxO1) is a key transcription factor that integrates insulin signaling with hepatic metabolism and becomes deregulated in the insulin-resistant liver, contributing to dyslipidemia in obesity. To gain mechanistic insights into how insulin resistance is linked to cholelithiasis, here we determined FoxO1's role in bile acid homeostasis and its contribution to cholelithiasis. We hypothesized that hepatic FoxO1 deregulation links insulin resistance to impaired bile acid metabolism and cholelithiasis. To address this hypothesis, we used the FoxO1LoxP/LoxP-Albumin-Cre system to generate liver-specific FoxO1-knockout mice. FoxO1-knockout mice and age- and sex-matched WT littermates were fed a lithogenic diet, and bile acid metabolism and gallstone formation were assessed in these animals. We showed that FoxO1 affected bile acid homeostasis by regulating hepatic expression of key enzymes in bile acid synthesis and in biliary cholesterol and phospholipid secretion. Furthermore, FoxO1 inhibited hepatic expression of the bile acid receptor farnesoid X receptor and thereby counteracted hepatic farnesoid X receptor signaling. Nonetheless, hepatic FoxO1 depletion neither affected the onset of gallstone disease nor impacted the disease progression, as FoxO1-knockout and control mice of both sexes had similar gallstone weights and incidence rates. These results argue against the notion that FoxO1 is a link between insulin resistance and cholelithiasis.
Collapse
Affiliation(s)
- Xiaoyun Feng
- Division of Endocrinology and Diabetes, Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224.,Department of Endocrinology & Metabolism, Shanghai General Hospital, Shanghai Jiaotong University, Shanghai 200080, China
| | - Cuiling Zhu
- Division of Endocrinology and Diabetes, Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224.,Department of Endocrinology & Metabolism, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Sojin Lee
- Division of Endocrinology and Diabetes, Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224
| | - Jingyang Gao
- Division of Endocrinology and Diabetes, Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224.,Department of Endocrinology & Metabolism, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Ping Zhu
- Division of Endocrinology and Diabetes, Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224.,Department of Endocrinology and Metabolism, Guangzhou Red Cross Hospital, Medical College of Jinan University, Guangzhou 510220, China
| | - Jun Yamauchi
- Division of Endocrinology and Diabetes, Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224
| | - Chenglin Pan
- Division of Endocrinology and Diabetes, Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224.,Department of Pediatrics, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Sucha Singh
- Division of Experimental Pathology, Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224.,Pittsburgh Liver Research Center, Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224
| | - Shen Qu
- Department of Endocrinology & Metabolism, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Rita Miller
- Division of Endocrinology and Diabetes, Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224
| | - Satdarshan P Monga
- Division of Experimental Pathology, Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224.,Pittsburgh Liver Research Center, Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224
| | - Yongde Peng
- Department of Endocrinology & Metabolism, Shanghai General Hospital, Shanghai Jiaotong University, Shanghai 200080, China
| | - H Henry Dong
- Division of Endocrinology and Diabetes, Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224 .,Pittsburgh Liver Research Center, Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15224
| |
Collapse
|
37
|
Adipose Tissue and FoxO1: Bridging Physiology and Mechanisms. Cells 2020; 9:cells9040849. [PMID: 32244542 PMCID: PMC7226803 DOI: 10.3390/cells9040849] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/23/2020] [Accepted: 03/30/2020] [Indexed: 12/22/2022] Open
Abstract
Forkhead box O class proteins (FoxOs) are expressed nearly in all tissues and are involved in different functions such as energy metabolism, redox homeostasis, differentiation, and cell cycle arrest. The plasticity of FoxOs is demonstrated by post-translational modifications that determine diverse levels of transcriptional regulations also controlled by their subcellular localization. Among the different members of the FoxO family, we will focus on FoxO1 in adipose tissue, where it is abundantly expressed and is involved in differentiation and transdifferentiation processes. The capability of FoxO1 to respond differently in dependence of adipose tissue subtype underlines the specific involvement of the transcription factor in energy metabolism and the “browning” process of adipocytes. FoxO1 can localize to nuclear, cytoplasm, and mitochondrial compartments of adipocytes responding to different availability of nutrients and source of reactive oxygen species (ROS). Specifically, fasted state produced-ROS enhance the nuclear activity of FoxO1, triggering the transcription of lipid catabolism and antioxidant response genes. The enhancement of lipid catabolism, in combination with ROS buffering, allows systemic energetic homeostasis and metabolic adaptation of white/beige adipocytes. On the contrary, a fed state induces FoxO1 to accumulate in the cytoplasm, but also in the mitochondria where it affects mitochondrial DNA gene expression. The importance of ROS-mediated signaling in FoxO1 subcellular localization and retrograde communication will be discussed, highlighting key aspects of FoxO1 multifaceted regulation in adipocytes.
Collapse
|
38
|
Tissue-Specific Metabolic Regulation of FOXO-Binding Protein: FOXO Does Not Act Alone. Cells 2020; 9:cells9030702. [PMID: 32182991 PMCID: PMC7140670 DOI: 10.3390/cells9030702] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/09/2020] [Accepted: 03/10/2020] [Indexed: 12/17/2022] Open
Abstract
The transcription factor forkhead box (FOXO) controls important biological responses, including proliferation, apoptosis, differentiation, metabolism, and oxidative stress resistance. The transcriptional activity of FOXO is tightly regulated in a variety of cellular processes. FOXO can convert the external stimuli of insulin, growth factors, nutrients, cytokines, and oxidative stress into cell-specific biological responses by regulating the transcriptional activity of target genes. However, how a single transcription factor regulates a large set of target genes in various tissues in response to a variety of external stimuli remains to be clarified. Evidence indicates that FOXO-binding proteins synergistically function to achieve tightly controlled processes. Here, we review the elaborate mechanism of FOXO-binding proteins, focusing on adipogenesis, glucose homeostasis, and other metabolic regulations in order to deepen our understanding and to identify a novel therapeutic target for the prevention and treatment of metabolic disorders.
Collapse
|
39
|
Nwadozi E, Rudnicki M, Haas TL. Metabolic Coordination of Pericyte Phenotypes: Therapeutic Implications. Front Cell Dev Biol 2020; 8:77. [PMID: 32117997 PMCID: PMC7033550 DOI: 10.3389/fcell.2020.00077] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 01/29/2020] [Indexed: 12/15/2022] Open
Abstract
Pericytes are mural vascular cells found predominantly on the abluminal wall of capillaries, where they contribute to the maintenance of capillary structural integrity and vascular permeability. Generally quiescent cells in the adult, pericyte activation and proliferation occur during both physiological and pathological vascular and tissue remodeling. A considerable body of research indicates that pericytes possess attributes of a multipotent adult stem cell, as they are capable of self-renewal as well as commitment and differentiation into multiple lineages. However, pericytes also display phenotypic heterogeneity and recent studies indicate that lineage potential differs between pericyte subpopulations. While numerous microenvironmental cues and cell signaling pathways are known to regulate pericyte functions, the roles that metabolic pathways play in pericyte quiescence, self-renewal or differentiation have been given limited consideration to date. This review will summarize existing data regarding pericyte metabolism and will discuss the coupling of signal pathways to shifts in metabolic pathway preferences that ultimately regulate pericyte quiescence, self-renewal and trans-differentiation. The association between dysregulated metabolic processes and development of pericyte pathologies will be highlighted. Despite ongoing debate regarding pericyte classification and their functional capacity for trans-differentiation in vivo, pericytes are increasingly exploited as a cell therapy tool to promote tissue healing and regeneration. Ultimately, the efficacy of therapeutic approaches hinges on the capacity to effectively control/optimize the fate of the implanted pericytes. Thus, we will identify knowledge gaps that need to be addressed to more effectively harness the opportunity for therapeutic manipulation of pericytes to control pathological outcomes in tissue remodeling.
Collapse
Affiliation(s)
| | | | - Tara L. Haas
- School of Kinesiology and Health Science, Angiogenesis Research Group and Muscle Health Research Centre, York University, Toronto, ON, Canada
| |
Collapse
|
40
|
Zou H, Hu R, Dong X, Shah AM, Wang Z, Ma J, Peng Q, Xue B, Wang L, Zhang X, Zeng S, Wang X, Shi J, Li F. Lipid Catabolism in Starved Yak Is Inhibited by Intravenous Infusion of β-Hydroxybutyrate. Animals (Basel) 2020; 10:ani10010136. [PMID: 31952136 PMCID: PMC7022817 DOI: 10.3390/ani10010136] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/06/2020] [Accepted: 01/11/2020] [Indexed: 12/30/2022] Open
Abstract
Simple Summary Yak, which is the predominant and semi-domesticated livestock on the Qinghai-Tibet Plateau, suffers severe starvation and body weight reduction in the cold season because of the harsh highland environment. Lipids are important energy sources to starvation animals. β-hydroxybutyrate (BHBA) that is derived from lipid decomposition as the primary ketone body is with the function not only to provide energy for animals as energy materials, but also regulate lipid metabolism as signaling molecular. However, the effects of starvation and BHBA on lipid metabolism and its mechanism are still unclear for ruminant animals. Herein, we investigated the effects of starvation and intravenous infusion of BHBA solution on Yak growth, serum biochemistry, hormones, subcutaneous adipocyte morphology, fatty acid composition, activity of enzymes related to lipid metabolism, and signal pathway. The results showed that starvation promoted lipid catabolism and BHBA infusion up-regulated the mRNA expression of receptor GPR109A in subcutaneous adipose tissue, inhibited the Cyclic adenosine monophosphate(cAMP)/Protein kinase A (PKA)/cAMP-responsive element binding protein (CREB) signaling pathway, and inhibited lipolysis. Our study was beneficial for enriching the nutrition regulation theory of yaks and improving their growth potential. Abstract Lipid is the chief energy source for starved animals. β-hydroxybutyrate (BHBA) is the main ketone body produced by lipid decomposition. In Chinese hamster ovary (CHO) cell experiment, it was found that BHBA could be used not only as an energy substance, but also as a ligand of GPR109A for regulating lipid metabolism. However, whether BHBA can regulate lipid metabolism of yaks, and its effective concentration and signal pathway are not clear. This study investigated the effects and mechanism of starvation and BHBA on the lipid metabolism of yak. Eighteen male Jiulong yaks were selected and then randomly divided into three groups: normal feeding group (NG), starvation group (SG), and starvation with BHBA infusion group (SBG). The yaks in the NG group were freely fed during the trial, while the yaks in the SG and SBG groups fasted; from 7th to 9th days of the experiment, the NG and SG were infused continuous with 0.9% normal saline and SBG was infused 1.7 mmol/L BHBA solution respectively. The blood samples were collected on the 0th, 1st, 3rd, 5th, 7th, and 9th day of experiment. The subcutaneous adipose tissue of all the yaks in this study were taken from live bodies after infusion. Serum glucose, lipid metabolites, hormone concentrations, and mRNA and protein expressions of key factors of lipid metabolism and signaling pathway in subcutaneous adipose tissue were measured. The results showed that, as compared with NG, starvation significantly reduced the body weight of yak in SG, and significantly increased the concentration of BHBA in serum and the mRNA expression of PKA and CREB1 in subcutaneous adipose tissue, while the mRNA expression of MEK, PKC, ERK1/2, the area of adipocytes, and the proportion of saturated fatty acid were decreased. Whereas, further increase of BHBA concentration through infusion promoted the mRNA expression of GPR109A receptor in the subcutaneous adipose tissue of SBG, inhibited the mRNA expression of AC and PKA, and decreased the phosphorylation protein abundance of CREB1, and significantly increased the diameter and area of adipocytes. These findings suggest that starvation led to enhanced lipid catabolism in yaks. An increasing BHBA concentration could increase the mRNA expression of GPR109A receptor in subcutaneous adipose tissue and inhibit the cAMP/PKA/CREB signaling pathway and lipid decomposition.
Collapse
Affiliation(s)
- Huawei Zou
- “Low Carbon Breeding Cattle and Safety Production” University Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 61130, China; (H.Z.); (R.H.); (X.D.); (A.M.S.); (J.M.); (Q.P.); (B.X.); (L.W.); (X.Z.); (S.Z.); (X.W.); (J.S.); (F.L.)
| | - Rui Hu
- “Low Carbon Breeding Cattle and Safety Production” University Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 61130, China; (H.Z.); (R.H.); (X.D.); (A.M.S.); (J.M.); (Q.P.); (B.X.); (L.W.); (X.Z.); (S.Z.); (X.W.); (J.S.); (F.L.)
| | - Xianwen Dong
- “Low Carbon Breeding Cattle and Safety Production” University Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 61130, China; (H.Z.); (R.H.); (X.D.); (A.M.S.); (J.M.); (Q.P.); (B.X.); (L.W.); (X.Z.); (S.Z.); (X.W.); (J.S.); (F.L.)
| | - Ali Mujtaba Shah
- “Low Carbon Breeding Cattle and Safety Production” University Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 61130, China; (H.Z.); (R.H.); (X.D.); (A.M.S.); (J.M.); (Q.P.); (B.X.); (L.W.); (X.Z.); (S.Z.); (X.W.); (J.S.); (F.L.)
- Department of Livestock Production, Shaheed Benazir Bhutto University of Veterinary and Animal Sciences, Sakrand 67210, Pakistan
| | - Zhisheng Wang
- “Low Carbon Breeding Cattle and Safety Production” University Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 61130, China; (H.Z.); (R.H.); (X.D.); (A.M.S.); (J.M.); (Q.P.); (B.X.); (L.W.); (X.Z.); (S.Z.); (X.W.); (J.S.); (F.L.)
- Correspondence:
| | - Jian Ma
- “Low Carbon Breeding Cattle and Safety Production” University Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 61130, China; (H.Z.); (R.H.); (X.D.); (A.M.S.); (J.M.); (Q.P.); (B.X.); (L.W.); (X.Z.); (S.Z.); (X.W.); (J.S.); (F.L.)
| | - Quanhui Peng
- “Low Carbon Breeding Cattle and Safety Production” University Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 61130, China; (H.Z.); (R.H.); (X.D.); (A.M.S.); (J.M.); (Q.P.); (B.X.); (L.W.); (X.Z.); (S.Z.); (X.W.); (J.S.); (F.L.)
| | - Bai Xue
- “Low Carbon Breeding Cattle and Safety Production” University Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 61130, China; (H.Z.); (R.H.); (X.D.); (A.M.S.); (J.M.); (Q.P.); (B.X.); (L.W.); (X.Z.); (S.Z.); (X.W.); (J.S.); (F.L.)
| | - Lizhi Wang
- “Low Carbon Breeding Cattle and Safety Production” University Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 61130, China; (H.Z.); (R.H.); (X.D.); (A.M.S.); (J.M.); (Q.P.); (B.X.); (L.W.); (X.Z.); (S.Z.); (X.W.); (J.S.); (F.L.)
| | - Xiangfei Zhang
- “Low Carbon Breeding Cattle and Safety Production” University Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 61130, China; (H.Z.); (R.H.); (X.D.); (A.M.S.); (J.M.); (Q.P.); (B.X.); (L.W.); (X.Z.); (S.Z.); (X.W.); (J.S.); (F.L.)
| | - Shaoyu Zeng
- “Low Carbon Breeding Cattle and Safety Production” University Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 61130, China; (H.Z.); (R.H.); (X.D.); (A.M.S.); (J.M.); (Q.P.); (B.X.); (L.W.); (X.Z.); (S.Z.); (X.W.); (J.S.); (F.L.)
| | - Xueying Wang
- “Low Carbon Breeding Cattle and Safety Production” University Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 61130, China; (H.Z.); (R.H.); (X.D.); (A.M.S.); (J.M.); (Q.P.); (B.X.); (L.W.); (X.Z.); (S.Z.); (X.W.); (J.S.); (F.L.)
| | - Junhua Shi
- “Low Carbon Breeding Cattle and Safety Production” University Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 61130, China; (H.Z.); (R.H.); (X.D.); (A.M.S.); (J.M.); (Q.P.); (B.X.); (L.W.); (X.Z.); (S.Z.); (X.W.); (J.S.); (F.L.)
| | - Fengpeng Li
- “Low Carbon Breeding Cattle and Safety Production” University Key Laboratory of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 61130, China; (H.Z.); (R.H.); (X.D.); (A.M.S.); (J.M.); (Q.P.); (B.X.); (L.W.); (X.Z.); (S.Z.); (X.W.); (J.S.); (F.L.)
| |
Collapse
|
41
|
Boughanem H, Cabrera-Mulero A, Millán-Gómez M, Garrido-Sánchez L, Cardona F, Tinahones FJ, Moreno-Santos I, Macías-González M. Transcriptional Analysis of FOXO1, C/EBP-α and PPAR-γ2 Genes and Their Association with Obesity-Related Insulin Resistance. Genes (Basel) 2019; 10:genes10090706. [PMID: 31547433 PMCID: PMC6770962 DOI: 10.3390/genes10090706] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 08/28/2019] [Accepted: 09/10/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Obesity is associated with several comorbid disorders, ranging from cardiovascular diseases to insulin resistance. In this context, visceral adipose tissue (VAT) seems to have a close connection with insulin resistance. In our study, we hypothesized that the expression profile of key adipogenic genes, such as proliferator-activated receptor γ type 2 (PPAR-γ2), CCAAT/enhancer-binding protein type α (C/EBP-α), and forkhead box protein class O type 1 (FOXO1) in VAT should shed light on their association with obesity-related insulin resistance. METHODS To test this idea, we studied the expression profile of C/EBP-α, FOXO1 and PPAR-γ2 in VAT from non-obese individuals, and low insulin (LIR-MO) and high insulin morbidly obese (HIR-MO) subjects, through a combination of RT-qPCR, co-immunoprecipitation, ELISA, Western blot analysis and EMSA assays. RESULTS Our results show that C/EBP-α and PPAR-γ2 were down-expressed in HIR-MO individuals, while FOXO1 was overexpressed. In addition, the PPAR-γ2-RXR-α heterodimer showed weak activity and bound weakly to the putative IGFBP-2-PPRE promoter sequence in VAT from HIR-MO subjects when compared with LIR-MO individuals. CONCLUSIONS These results show that PPAR-γ2, C/EBP-α, FOXO1 and IGFBP-2 have a close relationship with insulin resistance in VAT of morbidly obese individuals.
Collapse
Affiliation(s)
- Hatim Boughanem
- Biomedical Research Institute of Malaga (IBIMA), Faculty of Science, University of Malaga, 29010 Málaga, Spain.
| | - Amanda Cabrera-Mulero
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, University of Malaga (IBIMA), 29010 Málaga, Spain.
- CIBEROBN (CIBER in Physiopathology of Obesity and Nutrition CB06/03/0018), "Instituto de Salud Carlos III", 28029 Madrid, Spain.
| | - Mercedes Millán-Gómez
- CIBERCV (CIBER in cardiovascular diseases), "Instituto de Salud Carlos III", 28029 Madrid, Spain.
- Unidad de Gestión Clínica Área del Corazón, Virgen de la Victoria University Hospital, University of Malaga (IBIMA), 29010 Málaga, Spain.
| | - Lourdes Garrido-Sánchez
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, University of Malaga (IBIMA), 29010 Málaga, Spain.
- CIBEROBN (CIBER in Physiopathology of Obesity and Nutrition CB06/03/0018), "Instituto de Salud Carlos III", 28029 Madrid, Spain.
| | - Fernando Cardona
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, University of Malaga (IBIMA), 29010 Málaga, Spain.
- CIBEROBN (CIBER in Physiopathology of Obesity and Nutrition CB06/03/0018), "Instituto de Salud Carlos III", 28029 Madrid, Spain.
| | - Francisco José Tinahones
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, University of Malaga (IBIMA), 29010 Málaga, Spain.
- CIBEROBN (CIBER in Physiopathology of Obesity and Nutrition CB06/03/0018), "Instituto de Salud Carlos III", 28029 Madrid, Spain.
| | - Inmaculada Moreno-Santos
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, University of Malaga (IBIMA), 29010 Málaga, Spain.
| | - Manuel Macías-González
- Department of Endocrinology and Nutrition, Virgen de la Victoria University Hospital, University of Malaga (IBIMA), 29010 Málaga, Spain.
- CIBEROBN (CIBER in Physiopathology of Obesity and Nutrition CB06/03/0018), "Instituto de Salud Carlos III", 28029 Madrid, Spain.
| |
Collapse
|
42
|
Role of p110a subunit of PI3-kinase in skeletal muscle mitochondrial homeostasis and metabolism. Nat Commun 2019; 10:3412. [PMID: 31363081 PMCID: PMC6667496 DOI: 10.1038/s41467-019-11265-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 07/02/2019] [Indexed: 12/12/2022] Open
Abstract
Skeletal muscle insulin resistance, decreased phosphatidylinositol 3-kinase (PI3K) activation and altered mitochondrial function are hallmarks of type 2 diabetes. To determine the relationship between these abnormalities, we created mice with muscle-specific knockout of the p110α or p110β catalytic subunits of PI3K. We find that mice with muscle-specific knockout of p110α, but not p110β, display impaired insulin signaling and reduced muscle size due to enhanced proteasomal and autophagic activity. Despite insulin resistance and muscle atrophy, M-p110αKO mice show decreased serum myostatin, increased mitochondrial mass, increased mitochondrial fusion, and increased PGC1α expression, especially PCG1α2 and PCG1α3. This leads to enhanced mitochondrial oxidative capacity, increased muscle NADH content, and higher muscle free radical release measured in vivo using pMitoTimer reporter. Thus, p110α is the dominant catalytic isoform of PI3K in muscle in control of insulin sensitivity and muscle mass, and has a unique role in mitochondrial homeostasis in skeletal muscle. Diabetes is associated with decreased PI3K activation in skeletal muscle. Here, the authors show that p110a is the predominant PI3K subunit in muscle, and show that its ablation in muscle, but not ablation of p110beta, leads to insulin resistance, increased proteosomal and autophagic activity, and altered mitochondria homeostasis in mice.
Collapse
|
43
|
Zhang K, Yuan Q, Xie J, Yuan L, Wang Y. PPAR-γ activation increases insulin secretion independent of CASK in INS-1 cells. Acta Biochim Biophys Sin (Shanghai) 2019; 51:715-722. [PMID: 31168600 DOI: 10.1093/abbs/gmz052] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Indexed: 01/03/2023] Open
Abstract
Peroxisome proliferator-activated receptor-γ (PPAR-γ) is expressed in pancreatic β cells and is involved in insulin secretion. However, the precise mechanisms remain unclear. Calcium/calmodulin-dependent serine protein kinase (CASK), which plays a vital role in the anchoring of insulin granules on pancreatic β cell membrane, is probably a downstream of the transcription factor PPAR-γ. The aim of the present study was to investigate the correlation among PPAR-γ, CASK and insulin secretion. We found that rosiglitazone (RSG) had a positive effect on the expression of CASK and PPAR-γ in INS-1 cells as shown by real-time polymerase chain reaction (PCR) and western blot analysis, but did not change the cellular location of CASK as shown by immunofluorescence assay. Knockdown of PPAR-γ significantly attenuated the mRNA and protein expression levels of CASK. ChIP-qPCR and luciferase assays showed that PPAR-γ bound with the Cask promoter, and promoter activity of Cask was elevated by RSG. RSG significantly enhanced the insulin secretion with potassium stimulation, but did not alter the insulin content as shown by potassium-stimulated insulin secretion assay. In addition, with RSG pretreatment, knockdown of Cask did not significantly affect the PPAR-γ activation-mediated insulin secretion. Moreover, electron microscopy demonstrated that with RSG pretreatment, silence of Cask did not change the number of vesicles anchored on the cell membranes compared with those in siCask-treated cells. Overall, the present study identifies that CASK is one of the PPAR-γ downstream targets and PPAR-γ exerts a positive effect on the expression of CASK in INS-1 cells. PPAR-γ activation increases insulin secretion independent of the upregulation of CASK.
Collapse
Affiliation(s)
- Kai Zhang
- Department of Endocrinology, Zhongda Hospital, Institute of Diabetes, Medical School, Southeast University, Nanjing, China
| | - Qingzhao Yuan
- Department of Endocrinology, Zhongda Hospital, Institute of Diabetes, Medical School, Southeast University, Nanjing, China
| | - Jinyang Xie
- Department of Endocrinology, Zhongda Hospital, Institute of Diabetes, Medical School, Southeast University, Nanjing, China
| | - Li Yuan
- Department of Biochemistry and Molecular Biology, Nanjing Medical University, Nanjing, China
| | - Yao Wang
- Department of Endocrinology, Zhongda Hospital, Institute of Diabetes, Medical School, Southeast University, Nanjing, China
| |
Collapse
|
44
|
Analysis of differential gene expression of the transgenic pig with overexpression of PGC1α in muscle. Mol Biol Rep 2019; 46:3427-3435. [PMID: 30980266 DOI: 10.1007/s11033-019-04805-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 04/09/2019] [Indexed: 10/27/2022]
Abstract
In order to better understand the key regulatory mechanisms of PGC1α in muscle fiber type transition, the RNA-seq was used to compare the change of gene expression in gastrocnemius muscles between wild type pigs and transgenic pigs with overexpression of PGC1α gene in muscle. 371 differentially expressed genes (P ≤ 0.05 and Ratio ≥ 2), including 184 up-regulated genes and 187 down-regulated genes, were identified. Five main signaling pathways including metabolic pathways, ECM-receptor interaction, PPAR signaling pathway, adipocytokine signaling pathway and insulin signaling pathway, were authenticated using KEGG pathway analysis. Our results indicate that the fat metabolism pathway plays an important role in the transformation of muscle fiber types regulated by PGC1α.
Collapse
|
45
|
Sakaguchi M, Cai W, Wang CH, Cederquist CT, Damasio M, Homan EP, Batista T, Ramirez AK, Gupta MK, Steger M, Wewer Albrechtsen NJ, Singh SK, Araki E, Mann M, Enerbäck S, Kahn CR. FoxK1 and FoxK2 in insulin regulation of cellular and mitochondrial metabolism. Nat Commun 2019; 10:1582. [PMID: 30952843 PMCID: PMC6450906 DOI: 10.1038/s41467-019-09418-0] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 02/26/2019] [Indexed: 01/07/2023] Open
Abstract
A major target of insulin signaling is the FoxO family of Forkhead transcription factors, which translocate from the nucleus to the cytoplasm following insulin-stimulated phosphorylation. Here we show that the Forkhead transcription factors FoxK1 and FoxK2 are also downstream targets of insulin action, but that following insulin stimulation, they translocate from the cytoplasm to nucleus, reciprocal to the translocation of FoxO1. FoxK1/FoxK2 translocation to the nucleus is dependent on the Akt-mTOR pathway, while its localization to the cytoplasm in the basal state is dependent on GSK3. Knockdown of FoxK1 and FoxK2 in liver cells results in upregulation of genes related to apoptosis and down-regulation of genes involved in cell cycle and lipid metabolism. This is associated with decreased cell proliferation and altered mitochondrial fatty acid metabolism. Thus, FoxK1/K2 are reciprocally regulated to FoxO1 following insulin stimulation and play a critical role in the control of apoptosis, metabolism and mitochondrial function. Insulin signaling represses Forkhead transcription factor FoxO activity, which contributes to organismal metabolism. Here, the authors use proteomics to identify positively regulated insulin signaling targets FoxK1/K2 and demonstrate their role in lipid metabolism and mitochondrial regulation.
Collapse
Affiliation(s)
- Masaji Sakaguchi
- Sections of Integrative Physiology and Metabolism and Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center, Boston, MA, 02215, USA.,Department of Medicine, Harvard Medical School, Boston, MA, 02215, USA.,Department of Metabolic Medicine, Kumamoto University, 1-1-1 Honjo, Chuoku, Kumamoto, 860-8556, Japan
| | - Weikang Cai
- Sections of Integrative Physiology and Metabolism and Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center, Boston, MA, 02215, USA.,Department of Medicine, Harvard Medical School, Boston, MA, 02215, USA
| | - Chih-Hao Wang
- Sections of Integrative Physiology and Metabolism and Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center, Boston, MA, 02215, USA.,Department of Medicine, Harvard Medical School, Boston, MA, 02215, USA
| | - Carly T Cederquist
- Sections of Integrative Physiology and Metabolism and Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center, Boston, MA, 02215, USA.,Department of Medicine, Harvard Medical School, Boston, MA, 02215, USA
| | - Marcos Damasio
- Sections of Integrative Physiology and Metabolism and Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center, Boston, MA, 02215, USA.,Department of Medicine, Harvard Medical School, Boston, MA, 02215, USA
| | - Erica P Homan
- Sections of Integrative Physiology and Metabolism and Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center, Boston, MA, 02215, USA.,Department of Medicine, Harvard Medical School, Boston, MA, 02215, USA
| | - Thiago Batista
- Sections of Integrative Physiology and Metabolism and Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center, Boston, MA, 02215, USA.,Department of Medicine, Harvard Medical School, Boston, MA, 02215, USA
| | - Alfred K Ramirez
- Sections of Integrative Physiology and Metabolism and Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center, Boston, MA, 02215, USA.,Department of Medicine, Harvard Medical School, Boston, MA, 02215, USA
| | - Manoj K Gupta
- Sections of Integrative Physiology and Metabolism and Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center, Boston, MA, 02215, USA.,Department of Medicine, Harvard Medical School, Boston, MA, 02215, USA
| | - Martin Steger
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152, Martinsried, Germany
| | - Nicolai J Wewer Albrechtsen
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152, Martinsried, Germany.,Department of Biomedical Sciences and NNF Centre for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark.,Department of Clinical Proteomics, NNF Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Shailendra Kumar Singh
- Department of Host Defense, The World Premier International Research Center Initiative Immunology Frontier Research Center, Osaka, 565-0871, Japan
| | - Eiichi Araki
- Department of Metabolic Medicine, Kumamoto University, 1-1-1 Honjo, Chuoku, Kumamoto, 860-8556, Japan
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152, Martinsried, Germany
| | - Sven Enerbäck
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Medicinaregatan 9A, PO. Box. 440, 405 30, Göteborg, Sweden
| | - C Ronald Kahn
- Sections of Integrative Physiology and Metabolism and Islet Cell Biology and Regenerative Medicine, Joslin Diabetes Center, Boston, MA, 02215, USA. .,Department of Medicine, Harvard Medical School, Boston, MA, 02215, USA.
| |
Collapse
|
46
|
Chen J, Lu Y, Tian M, Huang Q. Molecular mechanisms of FOXO1 in adipocyte differentiation. J Mol Endocrinol 2019; 62:R239-R253. [PMID: 30780132 DOI: 10.1530/jme-18-0178] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 02/05/2019] [Indexed: 12/14/2022]
Abstract
Forkhead box-O1 (FOXO1) is a downstream target of AKT and plays crucial roles in cell cycle control, apoptosis, metabolism and adipocyte differentiation. It is thought that FOXO1 affects adipocyte differentiation by regulating lipogenesis and cell cycle. With the deepening in the understanding of this field, it is currently believed that FOXO1 translocation between nuclei and cytoplasm is involved in the regulation of FOXO1 activity, thus affecting adipocyte differentiation. Translocation of FOXO1 depends on its post-translational modifications and interactions with 14-3-3. Based on these modifications and interactions, FOXO1 could regulate lipogenesis through PPARγ and the adipocyte cell cycle through p21 and p27. In this review, we aim to provide a comprehensive FOXO1 regulation network in adipocyte differentiation by linking together distinct functions mentioned above to explain their effects on adipocyte differentiation and to emphasize the regulatory role of FOXO1. In addition, we also focus on the novel findings such as the use of miRNAs in FOXO1 regulation and highlight the improvable issues, such as RNA modifications, for future research in the field.
Collapse
Affiliation(s)
- Junye Chen
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, People's Republic of China
- Nanchang Joint Programme, Queen Mary, University of London, London, UK
| | - Yi Lu
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi Province, People's Republic of China
- Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, Jiangxi Province, People's Republic of China
| | - Mengyuan Tian
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, People's Republic of China
- Nanchang Joint Programme, Queen Mary, University of London, London, UK
| | - Qiren Huang
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang, Jiangxi Province, People's Republic of China
- Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang, Jiangxi Province, People's Republic of China
| |
Collapse
|
47
|
The immunosuppressant drug azathioprine restrains adipogenesis of muscle Fibro/Adipogenic Progenitors from dystrophic mice by affecting AKT signaling. Sci Rep 2019; 9:4360. [PMID: 30867438 PMCID: PMC6416262 DOI: 10.1038/s41598-019-39538-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 01/04/2019] [Indexed: 02/06/2023] Open
Abstract
Fibro/Adipogenic Progenitors (FAPs) define a stem cell population playing a pro-regenerative role after muscle damage. When removed from their natural niche, FAPs readily differentiate into adipocytes or fibroblasts. This digressive differentiation potential, which is kept under tight control in the healthy muscle niche, contributes to fat and scar infiltrations in degenerative myopathies, such as in Duchenne Muscular Dystrophy (DMD). Controlling FAP differentiation by means of small molecules may contribute to delay the adverse consequences of the progressive pathological degeneration while offering, at the same time, a wider temporal window for gene therapy and cell-based strategies. In a high content phenotypic screening, we identified the immunosuppressant, azathioprine (AZA) as a negative modulator of FAP adipogenesis. We show here that AZA negatively affects the adipogenic propensity of FAPs purified from wild type and mdx mice by impairing the expression of the master adipogenic regulator, peroxisome proliferator-activated receptor γ (PPARγ). We show that this inhibition correlates with a decline in the activation of the AKT-mTOR axis, the main pathway that transduces the pro-adipogenic stimulus triggered by insulin. In addition, AZA exerts a cytostatic effect that has a negative impact on the mitotic clonal process that is required for the terminal differentiation of the preadipocyte-committed cells.
Collapse
|
48
|
Tsuzuki K, Itoh Y, Inoue Y, Hayashi H. TRB
1 negatively regulates gluconeogenesis by suppressing the transcriptional activity of
FOXO
1. FEBS Lett 2019; 593:369-380. [DOI: 10.1002/1873-3468.13314] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 11/15/2018] [Accepted: 12/11/2018] [Indexed: 12/18/2022]
Affiliation(s)
- Kaori Tsuzuki
- Department of Cell Signaling Graduate School of Pharmaceutical Sciences Nagoya City University Japan
| | - Yuka Itoh
- Department of Cell Signaling Graduate School of Pharmaceutical Sciences Nagoya City University Japan
- Department of Biochemistry Graduate School of Medicine University of Yamanashi Japan
| | - Yasumichi Inoue
- Department of Cell Signaling Graduate School of Pharmaceutical Sciences Nagoya City University Japan
- Department of Innovative Therapeutics Sciences Cooperative Major in Nanopharmaceutical Sciences Graduate School of Pharmaceutical Sciences Nagoya City University Japan
| | - Hidetoshi Hayashi
- Department of Cell Signaling Graduate School of Pharmaceutical Sciences Nagoya City University Japan
- Department of Innovative Therapeutics Sciences Cooperative Major in Nanopharmaceutical Sciences Graduate School of Pharmaceutical Sciences Nagoya City University Japan
| |
Collapse
|
49
|
Zhou K, Yao P, He J, Zhao H. Lipophagy in nonliver tissues and some related diseases: Pathogenic and therapeutic implications. J Cell Physiol 2018; 234:7938-7947. [PMID: 30537019 DOI: 10.1002/jcp.27988] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 11/21/2018] [Indexed: 12/24/2022]
Abstract
Lipid autophagy (lipophagy) is defined as a selective autophagy process in which some intracellular lipid droplets are selectively degraded by autophagic lysosomes pathway. The occurrence of lipophagy was first discovered in liver tissues. Additionally, abundant evidence indicated that the occurrence of hepatic lipophagy has been implicated in many liver diseases including fatty liver diseases, nonalcoholic fatty liver diseases, liver fibrosis, and liver cirrhosis. However, recent studies suggested that hepatic lipophagy occurs not only in liver tissue but also in other nonliver tissues and cells. Furthermore, the occurrence of lipophagy plays a crucial role in nonliver tissues and some related diseases. For instance, lipophagy relieves insulin resistance in adipose tissue from obesity patient with type 2 diabetes. Additionally, lipophagy has the ability to remit neurodegenerative diseases by reducing activity-dependent neurodegeneration in nervous tissue. Lipophagy decreases muscle lipid accumulation and accordingly improves lipid storage myopathy in muscle tissue. Moreover, lipophagy alleviates the malignancy and metastasis of cancer in clear renal cell carcinoma tissue. Lipophagy is also involved in other processes, such as spermatogenesis, osteoblastogenesis, and mucosal ulceration. In conclusion, targeting lipophagy may be a critical regulator and a new therapeutic strategy for nonliver tissues and some related diseases.
Collapse
Affiliation(s)
- Kebing Zhou
- Department of Emergency Medicine, Affiliated Nanhua Hospital, University of South China, Hengyang, China
| | - Pingbo Yao
- Department of Emergency Medicine, Affiliated Nanhua Hospital, University of South China, Hengyang, China
| | - Jun He
- Department of Emergency Medicine, Affiliated Nanhua Hospital, University of South China, Hengyang, China
| | - Hong Zhao
- Department of Basic nursing, Nursing College, University of South China, Hengyang, China
| |
Collapse
|
50
|
Chen D, Gong Y, Xu L, Zhou M, Li J, Song J. Bidirectional regulation of osteogenic differentiation by the FOXO subfamily of Forkhead transcription factors in mammalian MSCs. Cell Prolif 2018; 52:e12540. [PMID: 30397974 PMCID: PMC6496202 DOI: 10.1111/cpr.12540] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 08/09/2018] [Accepted: 09/02/2018] [Indexed: 12/23/2022] Open
Abstract
Through loss‐ and gain‐of‐function experiments in knockout and transgenic mice, Forkhead box O (FOXO) family transcription factors have been demonstrated to play essential roles in many biological processes, including cellular proliferation, apoptosis and differentiation. Osteogenic differentiation from mesenchymal stem cells (MSCs) into osteoblasts is a well‐organized process that is carefully guided and characterized by various factors, such as runt‐related transcription factor 2 (Runx2), β‐catenin, osteocalcin (OCN), alkaline phosphatase (ALP) and activating transcription factor 4 (ATF4). Accumulating evidence suggests multiple interactions among FOXO members and the differentiation regulatory factors listed above, resulting in an enhancement or inhibition of osteogenesis in different stages of osteogenic differentiation. To systematically and integrally understand the role of FOXOs in osteogenic differentiation and explain the contrary phenomena observed in vitro and in vivo, we herein summarized FOXO‐interacting differentiation regulatory genes/factors and following alterations in differentiation. The underlying mechanism was further discussed on the basis of binding types, sites, phases and the consequent downstream transcriptional alterations of interactions among FOXOs and differentiation regulatory factors. Interestingly, a bidirectional effect of FOXOs on balancing osteogenic differentiation was discovered in MSCs. Moreover, FOXO factors are reported to be activated or suppressed by several context‐dependent signalling inputs during differentiation, and the underlying molecular basis may offer new drug development targets for treatments of bone formation defect diseases.
Collapse
Affiliation(s)
- Duanjing Chen
- College of Stomatology, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Yuanyuan Gong
- College of Stomatology, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Ling Xu
- College of Stomatology, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Mengjiao Zhou
- College of Stomatology, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Jie Li
- College of Stomatology, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Jinlin Song
- College of Stomatology, Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, Chongqing, China.,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| |
Collapse
|